DOI QR코드

DOI QR Code

HOXB13 is co-localized with androgen receptor to suppress androgen-stimulated prostate-specific antigen expression

  • Kim, Sin-Do (Department of Anatomy, Chonnam National University Medical School) ;
  • Park, Ra-Young (Department of Anatomy, Chonnam National University Medical School) ;
  • Kim, Young-Rang (Department of Anatomy, Chonnam National University Medical School) ;
  • Kim, In-Je (Department of Anatomy, Chonnam National University Medical School) ;
  • Kang, Taek-Won (Department of Urology, Chonnam National University Medical School) ;
  • Nam, Kwang-Il (Department of Anatomy, Chonnam National University Medical School) ;
  • Ahn, Kyu-Youn (Department of Anatomy, Chonnam National University Medical School) ;
  • Bae, Choon-Sang (Department of Anatomy, Chonnam National University Medical School) ;
  • Kim, Baik-Youn (Department of Anatomy, Chonnam National University Medical School) ;
  • Park, Sung-Sik (Department of Anatomy, Chonnam National University Medical School) ;
  • Jung, Chae-Yong (Department of Anatomy, Chonnam National University Medical School)
  • Received : 2010.07.14
  • Accepted : 2010.08.18
  • Published : 2010.12.30

Abstract

During the prostate cancer (PCa) development and its progression into hormone independency, androgen receptor (AR) signals play a central role by triggering the regulation of target genes, including prostate-specific antigen. However, the regulation of these AR-mediated target genes is not fully understood. We have previously demonstrated a unique role of HOXB13 homeodomain protein as an AR repressor. Expression of HOXB13 was highly restricted to the prostate and its suppression dramatically increased hormone-activated AR transactivation, suggesting that prostate-specific HOXB13 was a highly potent transcriptional regulator. In this report, we demonstrated the action mechanism of HOXB13 as an AR repressor. HOXB13 suppressed androgen-stimulated AR activity by interacting with AR. HOXB13 did neither bind to AR responsive elements nor disturb nuclear translocation of AR in response to androgen. In PCa specimen, we also observed mutual expression pattern of HOXB13 and AR. These results suggest that HOXB13 not only serve as a DNA-bound transcription factor but play an important role as an AR-interacting repressor to modulate hormone-activated androgen receptor signals. Further extensive studies will uncover a novel mechanism for regulating AR-signaling pathway to lead to expose new role of HOXB13 as a non-DNA-binding transcriptional repressor.

Keywords

Acknowledgement

Supported by : Chonnam National University

References

  1. Catron KM, Zhang H, Marshall SC, Inostroza JA, Wilson JM, Abate C. (1995). Transcriptional repression by Msx-1 does not require homeodomain DNA-binding sites. Mol Cell Biol 15: 861-871 https://doi.org/10.1128/MCB.15.2.861
  2. Chariot A, van Lint C, Chapelier M, Gielen J, Merville MP, Bours V. (1999). CBP and histone deacetylase inhibition enhance the transactivation potential of the HOXB7 homeodomain-containing protein. Oncogene 18: 4007-4014 https://doi.org/10.1038/sj.onc.1202776
  3. Economides KD, Capecchi MR. (2003). Hoxb13 is required for normal differentiation and secretory function of the ventral prostate. Development 130: 2061-2069 https://doi.org/10.1242/dev.00432
  4. Economides KD, Zeltser L, Capecchi MR. (2003). Hoxb13 mutations cause overgrowth of caudal spinal cord and tail vertebrae. Dev Biol 256: 317-330 https://doi.org/10.1016/S0012-1606(02)00137-9
  5. Edwards S, Campbell C, Flohr P, et al. (2005). Expression analysis onto microarrays of randomly selected cDNA clones highlights HOXB13 as a marker of human prostate cancer. Br J Cancer 92: 376-381 https://doi.org/10.1038/sj.bjc.6602261
  6. Hood L, Heath JR, Phelps ME, Lin B. (2004). Systems biology and new technologies enable predictive and preventative medicine. Science 306: 640-643 https://doi.org/10.1126/science.1104635
  7. Jung C, Kim RS, Lee SJ, Wang C, Jeng MH. (2004a). HOXB13 homeodomain protein suppresses the growth of prostate cancer cells by the negative regulation of T-cell factor 4. Cancer Res 64: 3046-3051 https://doi.org/10.1158/0008-5472.CAN-03-2614
  8. Jung C, Kim RS, Zhang HJ, Lee SJ, Jeng MH. (2004b). HOXB13 induces growth suppression of prostate cancer cells as a repressor of hormone-activated androgen receptor signaling. Cancer Res 64: 9185-9192 https://doi.org/10.1158/0008-5472.CAN-04-1330
  9. Kim YR, Oh KJ, Park RY, et al. (2010). HOXB13 promotes androgen independent growth of LNCaP prostate cancer cells by the activation of E2F signaling. Mol Cancer 9: 124 https://doi.org/10.1186/1476-4598-9-124
  10. Lagorce-Pages C, Paraf F, Dubois S, Belghiti J, Flejou JF. (1998). Expression of CD44 in premalignant and malignant Barrett's oesophagus. Histopathology 32: 7-14 https://doi.org/10.1046/j.1365-2559.1998.00316.x
  11. Lee SJ, Kim HS, Yu R, et al. (2002). Novel prostate-specific promoter derived from PSA and PSMA enhancers. Mol Ther 6: 415-421 https://doi.org/10.1006/mthe.2002.0682
  12. Lee SJ, Lee K, Yang X, et al. (2003). NFATc1 with AP-3 site binding specificity mediates gene expression of prostate-specific-membrane-antigen. J Mol Biol 330: 749-760 https://doi.org/10.1016/S0022-2836(03)00640-5
  13. Levine M, Hoey T. (1988). Homeobox proteins as sequence-specific transcription factors. Cell 55: 537-540 https://doi.org/10.1016/0092-8674(88)90209-7
  14. Mann RS, Chan SK. (1996). Extra specificity from extradenticle: the partnership between HOX and PBX/EXD homeodomain proteins. Trends Genet 12: 258-262 https://doi.org/10.1016/0168-9525(96)10026-3
  15. Podlasek CA, Clemens JQ, Bushman W. (1999a). Hoxa-13 gene mutation results in abnormal seminal vesicle and prostate development. J Urol 161: 1655-1661 https://doi.org/10.1016/S0022-5347(05)68999-9
  16. Podlasek CA, Duboule D, Bushman W. (1997). Male accessory sex organ morphogenesis is altered by loss of function of Hoxd-13. Dev Dyn 208: 454-465 https://doi.org/10.1002/(SICI)1097-0177(199704)208:4<454::AID-AJA2>3.0.CO;2-H
  17. Podlasek CA, Seo RM, Clemens JQ, Ma L, Maas RL, Bushman W. (1999b). Hoxa-10 deficient male mice exhibit abnormal development of the accessory sex organs. Dev Dyn 214: 1-12 https://doi.org/10.1002/(SICI)1097-0177(199901)214:1<1::AID-DVDY1>3.0.CO;2-2
  18. Prins GS, Birch L, Habermann H, et al. (2001). Influence of neonatal estrogens on rat prostate development. Reprod Fertil Dev 13: 241-252 https://doi.org/10.1071/RD00107
  19. Raman V, Martensen SA, Reisman D, et al. (2000a). Compromised HOXA5 function can limit p53 expression in human breast tumours. Nature 405: 974-978 https://doi.org/10.1038/35016125
  20. Raman V, Tamori A, Vali M, Zeller K, Korz D, Sukumar S. (2000b). HOXA5 regulates expression of the progesterone receptor. J Biol Chem 275: 26551-26555 https://doi.org/10.1074/jbc.C000324200
  21. Schnabel CA, Abate-Shen C. (1996). Repression by HoxA7 is mediated by the homeodomain and the modulatory action of its N-terminal-arm residues. Mol Cell Biol 16: 2678-2688 https://doi.org/10.1128/MCB.16.6.2678
  22. Shen W, Chrobak D, Krishnan K, Lawrence HJ, Largman C. (2004). HOXB6 protein is bound to CREB-binding protein and represses globin expression in a DNA binding-dependent, PBX interaction-independent process. J Biol Chem 279: 39895-39904 https://doi.org/10.1074/jbc.M404132200
  23. Shen WF, Chang CP, Rozenfeld S, et al. (1996). Hox homeodomain proteins exhibit selective complex stabilities with Pbx and DNA. Nucleic Acids Res 24: 898-906 https://doi.org/10.1093/nar/24.5.898
  24. Shen WF, Krishnan K, Lawrence HJ, Largman C. (2001). The HOX homeodomain proteins block CBP histone acetyltransferase activity. Mol Cell Biol 21: 7509-7522 https://doi.org/10.1128/MCB.21.21.7509-7522.2001
  25. Shen WF, Montgomery JC, Rozenfeld S, et al. (1997). AbdB-like Hox proteins stabilize DNA binding by the Meis1 homeodomain proteins. Mol Cell Biol 17: 6448-6458 https://doi.org/10.1128/MCB.17.11.6448
  26. Sreenath T, Orosz A, Fujita K, Bieberich CJ. (1999). Androgen-independent expression of hoxb-13 in the mouse prostate. Prostate 41: 203-207 https://doi.org/10.1002/(SICI)1097-0045(19991101)41:3<203::AID-PROS8>3.0.CO;2-J
  27. Takahashi Y, Hamada J, Murakawa K, et al. (2004). Expression profiles of 39 HOX genes in normal human adult organs and anaplastic thyroid cancer cell lines by quantitative real-time RT-PCR system. Exp Cell Res 293: 144-153 https://doi.org/10.1016/j.yexcr.2003.09.024
  28. Warot X, Fromental-Ramain C, Fraulob V, Chambon P, Dolle P. (1997). Gene dosage-dependent effects of the Hoxa-13 and Hoxd-13 mutations on morphogenesis of the terminal parts of the digestive and urogenital tracts. Development 124: 4781-4791
  29. Wimmel A, Kogan E, Ramaswamy A, Schuermann M. (2001). Variant expression of CD44 in preneoplastic lesions of the lung. Cancer 92: 1231-1236 https://doi.org/10.1002/1097-0142(20010901)92:5<1231::AID-CNCR1442>3.0.CO;2-Z
  30. Zappavigna V, Sartori D, Mavilio F. (1994). Specificity of HOX protein function depends on DNA-protein and protein-protein interactions, both mediated by the homeo domain. Genes Dev 8: 732-744 https://doi.org/10.1101/gad.8.6.732
  31. Zeltser L, Desplan C, Heintz N. (1996). Hoxb-13: a new Hox gene in a distant region of the HOXB cluster maintains colinearity. Development 122: 2475-2484

Cited by

  1. Confirmation of the HOXB13 G84E Germline Mutation in Familial Prostate Cancer vol.21, pp.8, 2010, https://doi.org/10.1158/1055-9965.epi-12-0495
  2. Dose-dependent effects of small-molecule antagonists on the genomic landscape of androgen receptor binding vol.13, pp.None, 2010, https://doi.org/10.1186/1471-2164-13-355
  3. HOXB13 Mutation and Prostate Cancer: Studies of Siblings and Aggressive Disease vol.22, pp.4, 2010, https://doi.org/10.1158/1055-9965.epi-12-1154
  4. HOX genes and their role in the development of human cancers vol.92, pp.8, 2010, https://doi.org/10.1007/s00109-014-1181-y
  5. HOXB13 regulates the prostate-derived Ets factor: Implications for prostate cancer cell invasion vol.45, pp.2, 2010, https://doi.org/10.3892/ijo.2014.2485
  6. HOXB13 downregulates intracellular zinc and increases NF-κB signaling to promote prostate cancer metastasis vol.33, pp.37, 2014, https://doi.org/10.1038/onc.2013.404
  7. Increased Expression of HOXB2 and HOXB13 Proteins is Associated with HPV Infection and Cervical Cancer Progression vol.16, pp.4, 2010, https://doi.org/10.7314/apjcp.2015.16.4.1349
  8. Prevalence of the HOXB13 G84E germline mutation in British men and correlation with prostate cancer risk, tumour characteristics and clinical outcomes vol.26, pp.4, 2010, https://doi.org/10.1093/annonc/mdv004
  9. Identification of Two Novel HOXB13 Germline Mutations in Portuguese Prostate Cancer Patients vol.10, pp.7, 2010, https://doi.org/10.1371/journal.pone.0132728
  10. Prostate Cancer Germline Variations and Implications for Screening and Treatment vol.8, pp.9, 2010, https://doi.org/10.1101/cshperspect.a030379
  11. Endogenous androgen receptor proteomic profiling reveals genomic subcomplex involved in prostate tumorigenesis vol.37, pp.3, 2010, https://doi.org/10.1038/onc.2017.330
  12. Prostate Organogenesis vol.8, pp.7, 2010, https://doi.org/10.1101/cshperspect.a030353
  13. Prostate Organogenesis vol.8, pp.7, 2010, https://doi.org/10.1101/cshperspect.a030353
  14. Targeting the BRD4-HOXB13 Coregulated Transcriptional Networks with Bromodomain-Kinase Inhibitors to Suppress Metastatic Castration-Resistant Prostate Cancer vol.17, pp.12, 2010, https://doi.org/10.1158/1535-7163.mct-18-0602
  15. The Homeobox gene, HOXB13, Regulates a Mitotic Protein-Kinase Interaction Network in Metastatic Prostate Cancers vol.9, pp.None, 2010, https://doi.org/10.1038/s41598-019-46064-4
  16. Prostate-specific markers to identify rare prostate cancer cells in liquid biopsies vol.16, pp.1, 2010, https://doi.org/10.1038/s41585-018-0119-5
  17. Genome-wide analysis of HOXC4 and HOXC6 regulated genes and binding sites in prostate cancer cells vol.15, pp.2, 2010, https://doi.org/10.1371/journal.pone.0228590
  18. Study on HOXBs of Clear Cell Renal Cell Carcinoma and Detection of New Molecular Target vol.2021, pp.None, 2010, https://doi.org/10.1155/2021/5541423