Skip to main content
Log in

Estrogen Metabolism as a Regulator of Estrogen Action in the Mammary Gland

  • Published:
Journal of Mammary Gland Biology and Neoplasia Aims and scope Submit manuscript

Abstract

Estrogen action in the target cells is dependent on estrogen receptor activity and intracellular estrogen concentration, which, in turn, is affected by the serum concentration and local metabolism in these cells. During the reproductive years the main source of estrogens is the ovarian follicles, but in postmenopausal women most of the estrogens are formed in peripheral tissues. 17β-hydroxysteroid dehydrogenases (17HSDs)6 catalyze the reaction between 17β-hydroxysteroids and 17-ketosteroids, and several distinct 17HSD isoenzymes have been characterized. 17HSD type 1 catalyzes the reaction from low-activity estrone to high-activity estradiol. The type 2 enzyme has an opposite activity, thereby reducing the exposure of tissues to estrogen action. 17HSD type 1 is expressed both in steroidogenic tissues and in the target tissues of steroid action, such as normal and malignant breast tissue, where it may be responsible for maintaining the high intracellular estradiol concentration seen in breast cancer specimens. Therefore, 17HSD type 1 inhibitors may be useful in the treatment and/or prevention of estrogen-dependent malignancies, such as breast cancer. This article deals mainly with 17HSD types 1 and 2 and their role in estrogen action in breast tissue.

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.

Similar content being viewed by others

REFERENCES

  1. J. S. Richards (1980). Maturation of ovarian follicles: Actions and interactions of pituitary and ovarian hormones on follicular cell differentiation. Physiol. Rev. 60:51–89.

    Google Scholar 

  2. J. Russo, B. A. Gusterson, A. E. Rogers, I. H. Russo, S. R. Wellings, and J. van Zwieten (1990). Comparative study of human and rat mammary tumorigenesis. Lab. Invest. 62:244–278.

    Google Scholar 

  3. G. J. Pepe and E. D. Albrecht (1995). Actions of placental and fetal adrenal steroid hormones in primate pregnancy. Endocrine Rev. 16:608–648.

    Google Scholar 

  4. E. Perel, M. E. Blackstein, and D. W. Killinger (1982). Aromatase in human breast carcinoma. Cancer Res. 42:3369s–3372s.

    Google Scholar 

  5. H. Sasano and N. Harada (1998). Intratumoral aromatase in human breast, endometrial, and ovarian malignancies. Endocrine Rev. 19:593–607.

    Google Scholar 

  6. J. R. Pasqualini, C. Celly, B.-L. Nguyen, and C. Vella (1989). Importance of estrogen sulfates in breast cancer. J. Steroid Biochem. 34:155–163.

    Google Scholar 

  7. S. J. Santner, P. D. Feil, and R. J. Santen (1984). In situ estrogen production via the estrone sulfatase pathway in breast tumors: Relative importance versus the aromatase pathway. J. Clin. Endocrinol. Metab. 59:29–33.

    Google Scholar 

  8. B. Longcope (1996). Dehydroepiandrosterone metabolism. J. Endocrinol. 150:S125–S127.

    Google Scholar 

  9. F. Labrie, V. Luu-The, S.-X. Lin, C. Labrie, J. Simard, R. Breton, and A. Bélanger (1997). The key role of 17β-hydroxysteroid dehydrogenases in sex steroid biology. Steroids 62:148–158.

    Google Scholar 

  10. A. Brodie, Q. Lu, and J. Nakamura (1997). Aromatase in the normal breast and breast cancer. J. Steroid Biochem. Mol. Biol. 61:281–286.

    Google Scholar 

  11. S. J. Santner, D. Leszczynski, C. Wright, A. Manni, P. D. Feil, and R. J. Santen (1986). Estrone sulfate: A potential source of estradiol in human breast cancer tissues. Breast Cancer Res. Treat. 7:35–44.

    Google Scholar 

  12. M. Poutanen, V. Isomaa, H. Peltoketo, and R. Vihko (1995). Role of 17β-hydroxysteroid dehydrogenase type 1 in endocrine and intracrine estradiol biosynthesis. J. Steroid Biochem. Mol. Biol. 55:525–532.

    Google Scholar 

  13. A. A. J. Van Landeghem, J. Poortman, M. Nabuurs, and J. H. H. Thijssen (1985). Endogenous concentration and subcellular distribution of estrogens in normal and malignant human breast tissue. Cancer Res. 45: 2900–2906.

    Google Scholar 

  14. A. Vermeulen, J. P. Deslypere, and R. Paridaens (1986). Steroid dynamics in the normal and carcinomatous mammary gland. J. Steroid Biochem. 25:799–802.

    Google Scholar 

  15. R. C. Bonney, M. J. Reed, P. A. Beranek, M. W. Ghilchik, and V. H. T. James (1986). Metabolism of [3H]oestradiol in vivo by normal breast and tumor tissue in postmenopausal women. J. Steroid Biochem. 24:361–364.

    Google Scholar 

  16. J. H. H. Thijssen, M. A. Blankenstein, W. R. Miller, and A. Milewicz (1987). Estrogens in tissues: Uptake from the peripheral circulation or local production. Steroids 50:297–306.

    Google Scholar 

  17. F. P. Li, J. A. Schneider, and A. F. Kantor (1993). Cancer epidemiology. Cancer Med. 1:332–339.

    Google Scholar 

  18. R. Vihko and D. Apter (1989). Endogenous steroids in the pathophysiology of breast cancer.CRC Crit. Rev Oncol/Hematol 9:1–16.

    Google Scholar 

  19. C. M. Mansfield (1993). A review of the etiology of breast cancer. J. Natl. Med. Assoc. 85:217–221.

    Google Scholar 

  20. L. Lipworth (1995). Epidemiology of breast cancer. Eur. J. Cancer. Prev. 4:7–30.

    Google Scholar 

  21. J. A. Cauley, J. P. Gutai, L. H. Kuller, D. LeDonne, and J. G. Powell (1989). The epidemiology of serum sex hormones in postmenopausal women. Am. J. Epidemiol. 129:1120–1131.

    Google Scholar 

  22. J. L. Kelsey and L. Bernstein (1996). Epidemiology and prevention of breast cancer. Ann. Rev. Publ. Health 17:47–67.

    Google Scholar 

  23. H. Spencer Feigelson and B. E. Henderson (1996). Estrogens and breast cancer. Carcinogenesis 17:2279–2284.

    Google Scholar 

  24. R. J. B. King (1991). A discussion of the roles of oestrogen and progestin in human mammary carcinogenesis. J. Steroid. Biochem. Mol. Biol. 39:811–818.

    Google Scholar 

  25. M. E. Lippman and R. B. Dickson (1989). Mechanisms of normal and malignant breast epithelial growth regulation. J. Steroid Biochem. 34:107–121.

    Google Scholar 

  26. R. J. Santen and H. Harvey (1999). Use of aromatase inhibitors in breast carcinoma Endocrine-Related Cancer 6:75–92.

    Google Scholar 

  27. H. Peltoketo, P. Vihko, and R. Vihko (1999). Regulation of estrogen action: Role of 17β-hydroxysteroid dehydrogenases. Vitamin Horm. 55:353–397.

    Google Scholar 

  28. N. Moghrabi, J. R. Head, and S. Andersson (1997). Cell typespecific expression of 17β-hydroxysteroid dehydrogenase type 2 in human placenta and fetal liver. J. Clin. Endocrinol. Metab. 82:3872–3878.

    Google Scholar 

  29. M. V. J. Mustonen, M. H. Poutanen, S. Kellokumpu, Y. de Launoit, V. V. Isomaa, R. K. Vihko, and P. T. Vihko (1998). Mouse 17β-hydroxysteroid dehydrogenase type 2 mRNA is predominantly expressed in hepatocytes and in surface epithelial cells of the gastrointestinal and urinary tracts. J. Mol. Endocrinol. 20:67–74.

    Google Scholar 

  30. H. Peltoketo, V. Luu-The, J. Simard, and J. Adamski (1999). 17β-Hydroxysteroid dehydrogenase (HSD)/17-ketosteroid reductase (KSR) family; Nomenclature and main characteristics of the 17HSD/KSR enzymes. J. Mol. Endocrinol. 23:1–11.

    Google Scholar 

  31. A. Krazeisen, R. Breitling, K. Imai, S. Fritz, G. Möller, and J. Adamski (1999). Determination of cDNA, gene structure and chromosomal localization of the novel human 17β-hydroxysteroid dehydrogenase type 7. FEBS Lett. 460:373–379.

    Google Scholar 

  32. Y.-M. Qin, M. H. Poutanen, H. M. Helander, A.-P. Kvist, K. M. Siivari, W. Schmitz, E. Conzelmann, U. Hellman, and K. Hiltunen (1997). Peroxisomal multifunctional enzyme of β-oxidation metabolizing D-3-hydroxyacyl-CoA esters in rat liver: Molecular cloning, expression and characterization. Biochem. J. 321:21–28.

    Google Scholar 

  33. S. A. Ghersevich, M. H. Poutanen, H. K. Martikainen, and R. K. Vihko (1994). Expression of 17β-hydroxysteroid dehydrogenase type 1 in human granulosa cells: Correlation with follicular size, cytochrome P450 aromatase activity and oestradiol production. J. Endocrinol. 143:139–150.

    Google Scholar 

  34. O. Mäentausta, R. Sormunen, V. Isomaa, V.-P. Lehto, P. Jouppila, and R. Vihko (1991). Immunohistochemical localization of 17β_-hydroxysteroid dehydrogenase in the human endometrium during the menstrual cycle. Lab. Invest. 65:582–587.

    Google Scholar 

  35. W. M. Geissler, D. L. Davis, L. Wu, K. D. Bradshaw, S. Patel, B. B. Mendoca, K. O. Elliston, J. D. Wilson, D. W. Russell, and S. Andersson (1994). Male pseudohermaphroditism caused by mutations of testicular 17β-hydroxysteroid dehydrogenase 3. Nat. Genet. 7:34–39.

    Google Scholar 

  36. J. A. Sha, K. Dudley, W. R. A. K. J. S. Rajapaksha, and P. J. O'shaughnessy (1997). Sequence of mouse 17β-hydroxysteroid dehydrogenase type 3 cDNA and tissue distribution of the type 1 and type 3 isoform mRNAs. J. Steroid Biochem. Mol. Biol. 60:19–24.

    Google Scholar 

  37. Y. Deyashiki, K. Ohshima, M. Nakanishi, K. Sato, K. Matsuura, and A. Hara (1995). Molecular cloning and characterization of mouse estradiol 17β-dehydrogenase (A-specific), a member of the aldoketoreductase family. J. Biol. Chem. 270:10461–10467.

    Google Scholar 

  38. M. G. Biswas and D. W. Russel (1997). Expression cloning and characterization of oxidative 17β-and 3β-hydroxysteroid dehydrogenases from rat and human prostate. J. Biol. Chem. 272:15959–15966.

    Google Scholar 

  39. P. Nokelainen, H. Peltoketo, R. Vihko, and P. Vihko (1998). Expression cloning of a novel estrogenic mouse 17β-hydroxysteroid dehydrogenase/17-ketosteroid reductase (m17HSD7), previously described as a prolactin receptor-associated protein (PRAP) in rat. Mol. Endocrinol. 12:1048–1059.

    Google Scholar 

  40. J. Fomitcheva, M. E. Baker, E. Anderson, G. Y. Lee, and N. Aziz (1998). Characterization of Ke 6, a new 17β-hydroxysteroid dehydrogenase, and its expression in gonadal tissues. J. Biol. Chem. 273:22664–22671.

    Google Scholar 

  41. M. L. Casey, P. C. MacDonald, and S. Andersson (1994). 17β-Hydroxysteroid dehydrogenase type 2: Chromosomal assignment and progestin regulation of gene expression in human endometrium. J. Clin. Invest. 94:2135–2141.

    Google Scholar 

  42. M. V. J. Mustonen, M. H. Poutanen, V. V. Isomaa, R. K. Vihko, and P. T. Vihko (1997). Cloning of mouse 17β-hydroxysteroid dehydrogenase type 2, and analyzing expression of themRNAs for types 1, 2, 3, 4, and 5 in mouse embryos and adult tissues. Biochem. J. 325:199–205.

    Google Scholar 

  43. M. V. J. Mustonen, V. V. Isomaa, T. Vaskivuo, J. Tapanainen, M. H. Poutanen, F. Stenbäck, R. K. Vihko, and P. T. Vihko (1998). Human 17β-hydroxysteroid dehydrogenase type 2 messenger ribonucleic acid expression and localization in term placenta and in endometrium during the menstrual cycle. J. Clin. Endocrinol. Metab. 83:1319–1324.

    Google Scholar 

  44. J. Adamski, T. Normand, F. Leenders, D. Monté, A. Begue, D. Stéhelin, P. W. Jungblut, and Y. de Launoit (1995). Molecular cloning of a novel widely expressed human 80 kDa 17β-hydroxysteroid dehydrogenase IV. Biochem. J. 311:437–443.

    Google Scholar 

  45. M. Poutanen, V. Isomaa, V.-P. Lehto, and R. Vihko (1992). Immunological analysis of 17β-hydroxysteroid dehydrogenase in benign and malignant human breast tissue. Int. J Cancer 50:386–390.

    Google Scholar 

  46. M. Poutanen, B. Moncharmont, and R. Vihko (1992). 17β-Hydroxysteroid dehydrogenase gene expression in human breast cancer cells: Regulation of expression by a progestin. Cancer Res. 52:290–294.

    Google Scholar 

  47. M. M. Miettinen, M. V. J. Mustonen, M. H. Poutanen, V. V. Isomaa, and R. K. Vihko (1996). Human 17β-hydroxysteroid dehydrogenase type 1 and type 2 isoenzymes have opposite activities in cultured cells and characteristic cell-and tissuespecific expression. Biochem. J. 314:839–845.

    Google Scholar 

  48. M. Poutanen, V. Isomaa, K. Kainulainen, and R. Vihko (1990). Progestin induction of 17β-hydroxysteroid dehydrogenase enzyme protein in the T-47D human breast-cancer cell line. Int. J. Cancer 46:897–901.

    Google Scholar 

  49. E. F. Adams, B. Rafferty, and M. C. White (1991). Interleukin 6 is secreted by breast fibroblasts and stimulates 17β-oestradiol oxidoreductase activity of MCF-7 cells: Possible paracrine regulation of breast 17β-oestradiol levels. Int. J. Cancer. 49:118–121.

    Google Scholar 

  50. L. J. Duncan, N. G. Goldham, and M. J. Reed (1994). The interaction of cytokines in regulating oestradiol 17β-hydroxysteroid dehydrogenase activity in MCF-7 cells. J. Steroid Biochem. Mol. Biol. 49:63–68.

    Google Scholar 

  51. A. Singh and M. J. Reed (1991). Insulin-like growth factor type I and insulin-like growth factor type II stimulate oestradiol-17β hydroxysteroid dehydrogenase (reductive) activity in breast cancer cells. J. Endocrinol. 129:R5–R8.

    Google Scholar 

  52. M. J. Reed, D. Rea, L. J. Duncan, and M. G. Parker (1994). Regulation of estradiol 17β-hydroxysteroid dehydrogenase expression and activity by retinoic acid in T47D breast cancer cells. Endocrinology 135:4–9.

    Google Scholar 

  53. Y.-S. Piao, H. Peltoketo, A. Jouppila, and R. Vihko (1997). Retinoic acids increase 17β-hydroxysteroid dehydrogenase type 1 expression in JEG-3 and T47D cells, but the stimulation is potentiated by epidermal growth factor, 12-O-tetradecanoylphorbol-13-acetate, and cyclic adenosine 3′,5′-monophosphate only in JEG-3 cells. Endocrinology 138:898–904.

    Google Scholar 

  54. M. M. Miettinen, M. H. Poutanen, and R. K. Vihko (1996). Characterization of estrogen-dependent growth of cultured MCF-7 human breast-cancer cells expressing 17β-hydroxysteroid dehydrogenase type 1. Int. J Cancer 68:600–604.

    Google Scholar 

  55. V. Luu The, C. Labrie, H. F. Zhao, J. Couët, Y. Lachance, J. Simard, G. Leblanc, J. Côté, D. Bérubé, R. Gagné, and F. Labrie (1989). Characterization of cDNAs for human estradiol 17β-dehydrogenase and assignment of the gene to chromosome 17: Evidence of two mRNA species with distinct 5′-termini in human placenta. Mol. Endocrinol. 3:1301–1309.

    Google Scholar 

  56. V. Luu-The, C. Labrie, J. Simard, Y. Lachance, H.-F. Zhao, J. Couët, G. Leblanc, and F. Labrie (1990). Structure of two in tandem human 17β-hydroxysteroid dehydrogenase genes. Mol. Endocrinol. 4:268–275.

    Google Scholar 

  57. H. Peltoketo, V. Isomaa, and R. Vihko (1992). Genomic organization and DNA sequences of human 17β-hydroxysteroid dehydrogenase dgenes and flanking regions. Eur. J. Biochem. 209:459–466.

    Google Scholar 

  58. Y. Tremblay, G. E. Ringler, Y. Morel, T. K. Mohandas, F. Labrie, J. F. Strauss, III, and W. L. Miller (1989). Regulation of the gene for estrogenic 17-ketosteroid reductase lying on chromosome 17cen→q25. J. Biol. Chem. 264:20458–20462.

    Google Scholar 

  59. Y.-S. Piao, H. Peltoketo, P. Vihko, and R. Vihko (1997). The proximal promoter region of the gene encoding human increase 17β-hydroxysteroid dehydrogenase type 1 contains GATA, AP-2 and Sp1 response elements: Analysis of promoter function in chorioncarcinoma cells. Endocrinology 138:3417–3425.

    Google Scholar 

  60. Y.-S. Piao, H. Peltoketo, J. Oikarinen, and R. Vihko (1995). Coordination of transcription of the human 17β-hydroxysteroid dehydrogenase type 1 gene (EDH17B2) by a cell-specific enhancer and a silencer: Identification of retinoic acid response element. Mol. Endocrinol. 9:1633–1644.

    Google Scholar 

  61. S. Leivonen, Y.-S. Piao, H. Peltoketo, P. Numchaisrika, R. Vihko, and P. Vihko (1999). Identification of essential subelements in the hHSD17B1 enhancer: Difference in function of the enhancer and that of the hHSD17BP1 analog is due to –480C and –486G. Endocrinology 140:3478–3487.

    Google Scholar 

  62. D. Ghosh, V. Z. Pletnev, D.-W. Zhu, Z. Wawrzak, W. L. Duax, W. Pangborn, F. Labrie, and S.-X. Lin (1995). Structure of human estrogenic 17β-hydroxysteroid dehydrogenase at 2. 20Å resolution. Structure 3:503–513.

    Google Scholar 

  63. M. Poutanen, M. Miettinen, and R. Vihko (1993). Differential estrogen specificities for transiently expressed human placental 17β-hydroxysteroid dehydrogenase and an endogenous enzyme expressed in cultured COS-m6 cells. Endocrinology 133:2639–2644.

    Google Scholar 

  64. T. Puranen, M. Poutanen, D. Ghosh, P. Vihko, and R. Vihko (1997). Characterization of structural and functional properties of human 17β-hydroxysteroid dehydrogenase type 1 using recombinant enzymes and site-directed mutants. Mol. Endocrinol. 11:77–86.

    Google Scholar 

  65. T. Puranen, M. Poutanen, D. Ghosh, R. Vihko, and P. Vihko (1997). Origin of substrate specificity of human and rat 17β-hydroxysteroid dehydrogenase type 1, using chimeric enzymes and site-directed substitutions. Endocrinology 138:3532–3539.

    Google Scholar 

  66. A. Azzi, P. H. Rehse, D. W. Zhu, R. L. Campabell, F. Labrie, and S.-X. Lin (1996). Crystal structure of human estrogenic 17β-hydroxysteroid dehydrogenase complexed with 17β-estradiol. Nat. Struct. Biol. 3(8):665–668.

    Google Scholar 

  67. R. Breton, D. Housset, C. Mazza, and J. C. Fontecill-Camps (1996). The structure of a complex of human 17β-hydroxysteroid dehydrogenase with estradiol and NADP+ identifies two principal targets for the design of inhibitors. Structure 4:905–915.

    Google Scholar 

  68. S.-X. Lin, D.-W. Zhu, A. Azzi, R. L. Campbell, R. Breton, F. Labrie, D. Ghosh, V. Pletnev, W. L. Duax, and W. Pangborn (1996). Studies on the three-dimensional structure of estrogenic 17β-hydroxysteroid dehydrogenase. J. Endocrinol. 150:S13–S20.

    Google Scholar 

  69. C. Mazza, R. Breton, D. Housset, and J. C. Fontecilla-Camps (1998). Unusual charge stabilization of NADP+ in 17β-hydroxysteroid dehydrogenase. J. Biol. Chem. 273:8145–8152.

    Google Scholar 

  70. H. Jörnvall, B. Persson, M. Krook, S. Atrian, R. Conzáles-Duarte, J. Jeffery, and D. Ghosh (1995). Short-chain dehydrogenases/reductases (SDR). Biochemistry 34:6003–6013.

    Google Scholar 

  71. D. Ghosh, C. M. Weeks, P. Grochulski W. L. Duax, M. Erman, R. L. Rimsay, and J. C. Orr (1991). Three-dimensional structure of holo 3α,20β-hydroxysteroid dehydrogenase:Amember of the short-chain dehydrogenase family. Proc. Natl. Acad. Sci. U.S.A. 88:10064–10068.

    Google Scholar 

  72. D. Ghosh, Z. Wawrzak, C. M. Weeks, W. L. Duax, and M. Erman (1994). The refined three-dimensional structure of 3α,20β-hydroxysteroid dehydrogenase and possible roles of the residues conserved in short-chain dehydrogenases. Structure 2:629–640.

    Google Scholar 

  73. T. M. Penning (1996). 17β-Hydroxysteroid dehydrogenase: Inhibitors and inhibitor design. Endocrine-Related Cancer 3:41–56.

    Google Scholar 

  74. M. W. Sawicki, M. Erman, T. Puranen, P. Vihko, and D. Ghosh (1999). Structure of the ternary complex of human 17β-hydroxysteroid dehydrogenase type 1 with 3-hydroxyestra-1,3,5,7-tetraen-17-one (equilin) and NADP+. Proc. Natl. Acad. Sci. U.S.A. 96:840–845.

    Google Scholar 

  75. M. W. Sawicki, N. Li, and D. Ghosh (1999). Equilin. Acta Crystallogr. 15:425–427.

    Google Scholar 

  76. L. Wu, M. Einstein, W. M. Geissler, H. K. Chan, K. O. Elliston, and S. Andersson (1993). Expression cloning and characterization of human 17β-hydroxysteroid dehydrogenase type 2, a microsomal enzyme possessing 20α-hydroxysteroid dehydrogenase activity. J. Biol. Chem. 268:12964–12969.

    Google Scholar 

  77. L. A. Akinola, M. Poutanen, and R. Vihko (1996). Cloning of rat 17β-hydroxysteroid dehydrogenase type 2 and characterization of tissue distribution and catalytic activity of rat type 1 and type 2 enzymes. Endocrinology 137:1572–1579.

    Google Scholar 

  78. J. P. Elo, L. A. Akinola, M. Poutanen, P. Vihko, A. P. Kyllö-nen, O. Lukkarinen, and R. Vihko (1996). Characterization of 17β-hydroxysteroid dehydrogenase isoenzyme expression in benign and malignant human prostate. Int. J. Cancer 66:37–41.

    Google Scholar 

  79. G. H. Tait, C. J. Newton, M. J. Reed, and V. H. T. James (1989). Multiple forms of 17β-hydroxysteroid oxidoreductase in human breast tissue. J. Mol. Endocrinol. 2:71–80.

    Google Scholar 

  80. V. Z. Mann, C. J. Newton, and G. H. Tait (1991). 17β-hydroxysteroid dehydrogenases in human breast tissues: Puri-fication and characterization of soluble enzymes and the distibution of particulate and soluble enzymes in adipose, nonadipose and tumor tissues. J. Mol. Endocrinol. 7:45–55.

    Google Scholar 

  81. M. Miettinen, M. Mustonen, M. Poutanen, V. Isomaa, M. Wickman, G. Söderqvist, R. Vihko, and P. Vihko (1999). 17β-Hydroxysteroid dehydrogenases in normal human mammary epithelial cells and breast tissue. Breast Cancer Res. Treat. 57:175–182.

    Google Scholar 

  82. W. R. Duan, D. I. H. Linzer, and G. Gibori (1996). Cloning and characterization of an ovarian specific protein that associates with the short form of the prolactin receptor. J. Biol. Chem. 271:15602–15607.

    Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Rights and permissions

Reprints and permissions

About this article

Cite this article

Miettinen, M., Isomaa, V., Peltoketo, H. et al. Estrogen Metabolism as a Regulator of Estrogen Action in the Mammary Gland. J Mammary Gland Biol Neoplasia 5, 259–270 (2000). https://doi.org/10.1023/A:1009542710520

Download citation

  • Issue Date:

  • DOI: https://doi.org/10.1023/A:1009542710520

Navigation