WNT/beta-catenin mediates radiation resistance of mouse mammary progenitor cells.
Journal: 2007/February - Proceedings of the National Academy of Sciences of the United States of America
ISSN: 0027-8424
Abstract:
Recent studies have identified a subpopulation of highly tumorigenic cells with stem/progenitor cell properties from human breast cancers, and it has been suggested that stem/progenitor cells, which remain after breast cancer therapy, may give rise to recurrent disease. We hypothesized that progenitor cells are resistant to radiation, a component of conventional breast cancer therapy, and that that resistance is mediated at least in part by Wnt signaling, which has been implicated in stem cell survival. To test this hypothesis, we investigated radioresistance by treating primary BALB/c mouse mammary epithelial cells with clinically relevant doses of radiation and found enrichment in normal progenitor cells (stem cell antigen 1-positive and side population progenitors). Radiation selectively enriched for progenitors in mammary epithelial cells isolated from transgenic mice with activated Wnt/beta-catenin signaling but not for background-matched controls, and irradiated stem cell antigen 1-positive cells had a selective increase in active beta-catenin and survivin expression compared with stem cell antigen 1-negative cells. In clonogenic assays, colony formation in the stem cell antigen 1-positive progenitors was unaffected by clinically relevant doses of radiation. Radiation also induced enrichment of side population progenitors in the human breast cancer cell line MCF-7. These data demonstrate that, compared with differentiated cells, progenitor cells have different cell survival properties that may facilitate the development of targeted antiprogenitor cell therapies.
Relations:
Content
Citations
(252)
References
(48)
Diseases
(1)
Chemicals
(5)
Genes
(1)
Organisms
(4)
Processes
(2)
Anatomy
(5)
Affiliates
(1)
Similar articles
Articles by the same authors
Discussion board
Proc Natl Acad Sci U S A 104(2): 618-623

WNT/β-catenin mediates radiation resistance of mouse mammary progenitor cells

*Department of Radiation Oncology, University of Texas M. D. Anderson Cancer Center, Houston, TX 77030; and
Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030
To whom correspondence should be addressed at: Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, M638a, Houston, TX 77030-3498., E-mail: ude.mcb@nesorj
Edited by Joan S. Brugge, Harvard Medical School, Boston, MA, and approved November 8, 2006

Author contributions: W.A.W. and M.S.C. contributed equally to this work; W.A.W., M.S.C., and J.M.R. designed research; W.A.W., M.S.C., F.B., and M.P.A. performed research; J.M.R. contributed new reagents/analytic tools; W.A.W., M.S.C., F.B., M.P.A., T.A.B., and J.M.R. analyzed data; and W.A.W., M.S.C., T.A.B., and J.M.R. wrote the paper.

Edited by Joan S. Brugge, Harvard Medical School, Boston, MA, and approved November 8, 2006
Received 2006 Aug 1

Abstract

Recent studies have identified a subpopulation of highly tumorigenic cells with stem/progenitor cell properties from human breast cancers, and it has been suggested that stem/progenitor cells, which remain after breast cancer therapy, may give rise to recurrent disease. We hypothesized that progenitor cells are resistant to radiation, a component of conventional breast cancer therapy, and that that resistance is mediated at least in part by Wnt signaling, which has been implicated in stem cell survival. To test this hypothesis, we investigated radioresistance by treating primary BALB/c mouse mammary epithelial cells with clinically relevant doses of radiation and found enrichment in normal progenitor cells (stem cell antigen 1-positive and side population progenitors). Radiation selectively enriched for progenitors in mammary epithelial cells isolated from transgenic mice with activated Wnt/β-catenin signaling but not for background-matched controls, and irradiated stem cell antigen 1-positive cells had a selective increase in active β-catenin and survivin expression compared with stem cell antigen 1-negative cells. In clonogenic assays, colony formation in the stem cell antigen 1-positive progenitors was unaffected by clinically relevant doses of radiation. Radiation also induced enrichment of side population progenitors in the human breast cancer cell line MCF-7. These data demonstrate that, compared with differentiated cells, progenitor cells have different cell survival properties that may facilitate the development of targeted antiprogenitor cell therapies.

Keywords: stem cell antigen 1, survivin, MCF-7, linCD24CD29, side population
Abstract

It has been speculated that stem cells may represent the cellular origins of cancer because they exist quiescently for long periods of time and could accumulate multiple mutations over the lifespan of an organism, ultimately giving rise to tumors when stimulated to proliferate (1). Recently, it was reported that highly tumorigenic cells with properties consistent with those of stem/progenitor cells can be isolated from human breast cancers (2). These data suggest that cancer stem cells may exist in human breast cancer and that they may have different biologic features than other, more differentiated cells that constitute the majority of the cells in human breast cancers. Conceivably, cancer stem cells may be more resistant to conventional breast cancer therapies, which may ultimately result in recurrence or metastasis even when remarkable initial responses are observed clinically (3).

Although the elucidation of sensitive stem and progenitor cell markers in the mammary gland and in breast tumors has been relatively recent, the normal tissue response of stem and progenitor cells to radiation, an integral component of multidisciplinary breast cancer therapy, has been the subject of several decades of radiobiological data. The development of a simple, reproducible in vivo clonogenic assay in the jejunum, where the effects of radiation on stem cells can be easily measured, led to the conclusions that, in the small intestine, the cells at position 4–5 in the crypt now identified as the stem cells are exquisitely sensitive to radiation (4). However, a second population of potential stem cells, elsewhere called transiently amplifying cells or progenitors, exists that can be called into action in the event of lethal damage to the stem cells. At low doses of radiation the crypt of the small intestine contains four to five clonogenic regenerating cells, but at higher doses of radiation up to 30–40 potential clonogenic regenerators can be called into play (4). The elucidation of stem/progenitor cell markers in the mammary gland now allow this work to be tested at the cellular level in the mammary gland. We hypothesize that mammary gland progenitors may be resistant to radiation and that this resistance is mediated by the β-catenin stem cell survival signaling pathway.

β-Catenin is an essential component of both intercellular junctions and the canonical Wnt signaling pathway, which has been implicated in stem cell survival (5). In recent studies, activation of β-catenin in granulocyte–macrophage progenitors in chronic myelogenous leukemia appeared to enhance their self-renewal activity and leukemic potential (3). In addition, studies in the intestine and mammary gland have linked β-catenin signaling to stem cell survival and tumorigenesis (68).

Several methods are currently being used to isolate and study mammary stem/progenitor cells, including long-term bromodeoxyuridine labeling to identify label-retaining cells, Hoechst dye efflux to identify side population (SP) properties, and the potential stem/progenitor cell–cell surface markers, such as stem cell antigen 1 (Sca1) and α6- and β1-integrins (911). In the hematopoietic system, cells that efflux Hoechst 33342 dye have been shown to comprise a small fraction of bone marrow, which are capable of recapitulating the bone marrow in irradiated mice, establishing their functional capacity as hematopoietic stem cells (12). These cells are represented as a SP on flow cytometry analysis present in both mouse and human mammary glands (1316). In hematopoietic stem cells, the efflux of Hoechst dye is due to the presence of a family of drug-effluxing protein pumps, including the breast cancer resistance protein-1 (BCRP1)/ABCG2 transporter, that may be responsible for drug resistance in many types of cancer (17). In the mouse mammary gland, the SP phenotype depends on several members of the ABC transporter family (18). Outgrowth experiments using SP are confounded by the toxicity of the Hoechst dye (14); however, the SP fraction in mouse mammary gland is enriched for long-term bromodeoxyuridine label-retaining cells, as well as Sca1-positive cells, which have the capacity to generate functional mammary outgrowths in transplantation experiments (13). Shackleton et al. (10) recently confirmed outgrowth potential of a Sca1 population but demonstrated that the majority of outgrowth potential of the Sca1 population in fact lies in the small, Sca1-positive cells. Taken together, these data support the SP and Sca1 phenotypes as useful surrogates for stem-like/progenitor cells. A thoughtful review of the mammary SP phenotype was published recently (19), and review of the literature regarding both SP and Sca1 suggests that each of these represents markers useful for isolating potential downstream progenitors (9) rather than the more primitive stem cells isolated as described by Shackleton et al. (10) as linCD24CD29.

Although it has been speculated that stem or progenitor cells in the mammary gland are more resistant to conventional cancer therapies, this relationship has not been explicitly demonstrated. Here we demonstrate that progenitors in murine mammary epithelial cell (MEC) culture are enriched by clinically relevant doses of ionizing radiation and that this enrichment is enhanced by β-catenin stabilization. In human breast cancer MCF-7 cells, radiation induced enrichment of both SP progenitors and a linCD24CD29 subpopulation. These findings from normal and genetically manipulated mouse mammary glands may have important implications for the evaluation of new and current cancer therapies and the development of future new anti-stem-cell-targeted therapies.

Click here to view.

Acknowledgments

We thank the Baylor College of Medicine Flow Core Laboratory and the Texas Children's Hospital Flow Laboratory for flow cytometry assistance. We acknowledge Frances Kittrell and Jessica Li for assistance. We also thank Drs. Daniel Medina and Peggy Goodell for their advice and helpful criticisms. This work is supported in part by National Cancer Institute Grant CA-16303 (to J.M.R.) and Department of Defense Grant Predoctoral Fellowship DAMD 17-03-1-0699 (to M.S.C.).

Acknowledgments

Abbreviations

MECmammary epithelial cell
SPside population
%SPpercent SP
Sca1stem cell antigen 1
PEphycoerythrin.
Abbreviations

Footnotes

The authors declare no conflict of interest.

This article is a PNAS direct submission.

This article contains supporting information online at www.pnas.org/cgi/content/full/0606599104/DC1.

Footnotes

References

  • 1. Sell S. Crit Rev Oncol Hematol. 2004;51:1–28.[PubMed]
  • 2. Al-Hajj M, Wicha MS, Benito-Hernandez A, Morrison SJ, Clarke MF. Proc Natl Acad Sci USA. 2003;100:3983–3988.
  • 3. Jamieson CH, Ailles LE, Dylla SJ, Muijtjens M, Jones C, Zehnder JL, Gotlib J, Li K, Manz MG, Keating A, et al N Engl J Med. 2004;351:657–667.[PubMed][Google Scholar]
  • 4. Potten CS. Radiat Res. 2004;161:123–136.[PubMed]
  • 5. Polakis P. Genes Dev. 2000;14:1837–1851.[PubMed]
  • 6. Theodosiou NA, Tabin CJ. Dev Biol. 2003;259:258–271.[PubMed]
  • 7. Taipale J, Beachy PA. Nature. 2001;411:349–354.[PubMed]
  • 8. Li Y, Welm B, Podsypanina K, Huang S, Chamorro M, Zhang X, Rowlands T, Egeblad M, Cowin P, Werb Z, et al Proc Natl Acad Sci USA. 2003;100:15853–15858.[Google Scholar]
  • 9. Woodward WA, Chen MS, Behbod F, Rosen JM. J Cell Sci. 2005;118:3585–3594.[PubMed]
  • 10. Shackleton M, Vaillant F, Simpson KJ, Stingl J, Smyth GK, Asselin-Labat ML, Wu L, Lindeman GJ, Visvader JE. Nature. 2006;439:84–88.[PubMed]
  • 11. Stingl J, Eirew P, Ricketson I, Shackleton M, Vaillant F, Choi D, Li HI, Eaves CJ. Nature. 2006;439:993–997.[PubMed]
  • 12. Goodell MA, Brose K, Paradis G, Conner AS, Mulligan RC. J Exp Med. 1996;183:1797–1806.
  • 13. Welm BE, Tepera SB, Venezia T, Graubert TA, Rosen JM, Goodell MA. Dev Biol. 2002;245:42–56.[PubMed]
  • 14. Alvi AJ, Clayton H, Joshi C, Enver T, Ashworth A, Vivanco MM, Dale TC, Smalley MJ. Breast Cancer Res. 2003;5:R1–R8.
  • 15. Clayton H, Titley I, Vivanco M. Exp Cell Res. 2004;297:444–460.[PubMed]
  • 16. Clarke RB, Spence K, Anderson E, Howell A, Okano H, Potten CS. Dev Biol. 2005;277:443–456.[PubMed]
  • 17. Hirschmann-Jax C, Foster AE, Wulf GG, Nuchtern JG, Jax TW, Gobel U, Goodell MA, Brenner MK. Proc Natl Acad Sci USA. 2004;101:14228–14233.
  • 18. Jonker JW, Freeman J, Bolscher E, Musters S, Alvi AJ, Titley I, Schinkel AH, Dale TC. Stem Cells. 2005;23:1059–1065.[PubMed]
  • 19. Smalley MJ, Clarke RB. J Mamm Gland Biol Neoplasia. 2005;10:37–47.[PubMed]
  • 20. Kondo T, Setoguchi T, Taga T. Proc Natl Acad Sci USA. 2004;101:781–786.
  • 21. Patrawala L, Calhoun T, Schneider-Broussard R, Zhou J, Claypool K, Tang DG. Cancer Res. 2005;65:6207–6219.[PubMed]
  • 22. Hall EJ Radiobiology for the Radiologist. Philadelphia: Lippincott, Williams & Wilkins; 2000. [PubMed][Google Scholar]
  • 23. Burma S, Chen BP, Murphy M, Kurimasa A, Chen DJ. J Biol Chem. 2001;276:42462–42467.[PubMed]
  • 24. Liu BY, McDermott SP, Khwaja SS, Alexander CM. Proc Natl Acad Sci USA. 2004;101:4158–4163.
  • 25. Okayasu R, Suetomi K, Yu Y, Silver A, Bedford JS, Cox R, Ullrich RL. Cancer Res. 2000;60:4342–4345.[PubMed]
  • 26. Storer JB, Mitchell TJ, Fry RJ. Radiat Res. 1988;114:331–353.[PubMed]
  • 27. Harada N, Tamai Y, Ishikawa T, Sauer B, Takaku K, Oshima M, Taketo MM. EMBO J. 1999;18:5931–5942.
  • 28. Kim PJ, Plescia J, Clevers H, Fearon ER, Altieri DC. Lancet. 2003;362:205–209.[PubMed]
  • 29. Yang D, Welm A, Bishop JM. Proc Natl Acad Sci USA. 2004;101:15100–15105.
  • 30. Bao S, Wu Q, McLendon RE, Hao Y, Shi Q, Hjelmeland AB, Dewhirst MW, Bigner DD, Rich JN. Nature. 2006;444:756–760.[PubMed]
  • 31. Deugnier MA, Faraldo MM, Teuliere J, Thiery JP, Medina D, Glukhova MA. Dev Biol. 2006;293:414–425.[PubMed]
  • 32. Sleeman KE, Kendrick H, Ashworth A, Isacke CM, Smalley MJ. Breast Cancer Res. 2006;8:R7.
  • 33. Pardal R, Clarke MF, Morrison SJ. Nat Rev Cancer. 2003;3:895–902.[PubMed]
  • 34. Lee HY, Kleber M, Hari L, Brault V, Suter U, Taketo MM, Kemler R, Sommer L. Science. 2004;303:1020–1023.[PubMed]
  • 35. Huelsken J, Birchmeier W. Curr Opin Genet Dev. 2001;11:547–553.[PubMed]
  • 36. Reya T, Duncan AW, Ailles L, Domen J, Scherer DC, Willert K, Hintz L, Nusse R, Weissman IL. Nature. 2003;423:409–414.[PubMed]
  • 37. van de Wetering M, Sancho E, Verweij C, de Lau W, Oving I, Hurlstone A, van der Horn K, Batlle E, Coudreuse D, Haramis AP, et al Cell. 2002;111:241–250.[PubMed][Google Scholar]
  • 38. Tepera SB, McCrea PD, Rosen JM. J Cell Sci. 2003;116:1137–1149.[PubMed]
  • 39. Emami KH, Nguyen C, Ma H, Kim DH, Jeong KW, Eguchi M, Moon RT, Teo JL, Kim HY, Moon SH, et al Proc Natl Acad Sci USA. 2004;101:12682–12687.[Google Scholar]
  • 40. Kim KM, Calabrese P, Tavare S, Shibata D. Am J Pathol. 2004;164:1369–1377.
  • 41. Endoh T, Tsuji N, Asanuma K, Yagihashi A, Watanabe N. Exp Cell Res. 2005;305:300–311.[PubMed]
  • 42. Wong SC, Lo SF, Lee KC, Yam JW, Chan JK, Wendy Hsiao WL. J Pathol. 2002;196:145–153.[PubMed]
  • 43. Klopocki E, Kristiansen G, Wild PJ, Klaman I, Castanos-Velez E, Singer G, Stohr R, Simon R, Sauter G, Leibiger H, et al Int J Oncol. 2004;25:641–649.[PubMed][Google Scholar]
  • 44. Lin SY, Xia W, Wang JC, Kwong KY, Spohn B, Wen Y, Pestell RG, Hung MC. Proc Natl Acad Sci USA. 2000;97:4262–4266.
  • 45. Jain M, Arvanitis C, Chu K, Dewey W, Leonhardt E, Trinh M, Sundberg CD, Bishop JM, Felsher DW. Science. 2002;297:102–104.[PubMed]
  • 46. Ayyanan A, Civenni G, Ciarloni L, Morel C, Mueller N, Lefort K, Mandinova A, Raffoul W, Fiche M, Dotto GP, Brisken C. Proc Natl Acad Sci USA. 2006;103:3799–3804.
  • 47. Behbod F, Rosen JM. Carcinogenesis. 2005;26:703–711.[PubMed]
  • 48. Anton M, Graham FL. J Virol. 1995;69:4600–4606.
  • 49. Pullan S, Wilson J, Metcalfe A, Edwards GM, Goberdhan N, Tilly J, Hickman JA, Dive C, Streuli CH. J Cell Sci. 1996;109:631–642.[PubMed]
Collaboration tool especially designed for Life Science professionals.Drag-and-drop any entity to your messages.