Semin Reprod Med 2015; 33(05): 357-365
DOI: 10.1055/s-0035-1558451
Thieme Medical Publishers 333 Seventh Avenue, New York, NY 10001, USA.

Uterine Leiomyoma Stem Cells: Linking Progesterone to Growth

Serdar E. Bulun
1   Department of Obstetrics and Gynecology, Feinberg School of Medicine at Northwestern University, Chicago, Illinois
,
Molly B. Moravek
1   Department of Obstetrics and Gynecology, Feinberg School of Medicine at Northwestern University, Chicago, Illinois
,
Ping Yin
1   Department of Obstetrics and Gynecology, Feinberg School of Medicine at Northwestern University, Chicago, Illinois
,
Masanori Ono
2   Department of Obstetrics and Gynecology, Keio University School of Medicine, Tokyo, Japan
,
John S. Coon V
1   Department of Obstetrics and Gynecology, Feinberg School of Medicine at Northwestern University, Chicago, Illinois
,
Matthew T. Dyson
1   Department of Obstetrics and Gynecology, Feinberg School of Medicine at Northwestern University, Chicago, Illinois
,
Antonia Navarro
1   Department of Obstetrics and Gynecology, Feinberg School of Medicine at Northwestern University, Chicago, Illinois
,
Erica E. Marsh
1   Department of Obstetrics and Gynecology, Feinberg School of Medicine at Northwestern University, Chicago, Illinois
,
Hong Zhao
1   Department of Obstetrics and Gynecology, Feinberg School of Medicine at Northwestern University, Chicago, Illinois
,
Tetsuo Maruyama
2   Department of Obstetrics and Gynecology, Keio University School of Medicine, Tokyo, Japan
,
Debabrata Chakravarti
1   Department of Obstetrics and Gynecology, Feinberg School of Medicine at Northwestern University, Chicago, Illinois
,
J. Julie Kim
1   Department of Obstetrics and Gynecology, Feinberg School of Medicine at Northwestern University, Chicago, Illinois
,
Jian-Jun Wei
1   Department of Obstetrics and Gynecology, Feinberg School of Medicine at Northwestern University, Chicago, Illinois
3   Department of Pathology, Feinberg School of Medicine at Northwestern University, Chicago, Illinois
› Author Affiliations
Further Information

Publication History

Publication Date:
06 August 2015 (online)

Abstract

Uterine leiomyomas (fibroids) represent the most common class of benign tumors in women. Multiple leiomyomas usually arise from the uterus of a symptomatic woman. These tumors cause a variety of symptoms, including abnormal uterine bleeding, pelvic pain, bladder or bowel dysfunction, and recurrent pregnancy loss, and are responsible for more than 200,000 hysterectomies in the United States annually. Each leiomyoma seems to arise from the clonal expansion of a single myometrial smooth muscle cell transformed by a mutation. Tumor expansion is sustained by cell proliferation together with the production of large amounts of extracellular matrix. Estrogen and progesterone stimulate the growth of leiomyomas. Estrogen, together with its receptor ERα, enables progesterone action via induction of progesterone receptor (PR) expression. Progesterone induces the growth of leiomyoma by regulation of a set of key genes that control proliferation and apoptosis. A distinct cell population with stem-progenitor properties is indispensable for progesterone-dependent growth of leiomyomas. This stem-progenitor cell population is deficient in ERα and PR and dependent on the much higher levels of these steroid receptors in surrounding mature leiomyoma or myometrial cells. Progesterone sends paracrine signals from these mature cells to stem cells. The WNT/β-catenin pathway comprises a key component of this paracrine signaling system. The majority of medical treatments currently available for leiomyoma works by inhibiting estrogen or progesterone production or action, but tumors tend to regrow once treatment is stopped. Targeting stem cells and their paracrine interactions with more differentiated cell populations within leiomyoma may lead to the development of more effective therapeutics.

 
  • References

  • 1 Parker WH. Etiology, symptomatology, and diagnosis of uterine myomas. Fertil Steril 2007; 87 (4) 725-736
  • 2 Myers ER, Barber MD, Gustilo-Ashby T, Couchman G, Matchar DB, McCrory DC. Management of uterine leiomyomata: what do we really know?. Obstet Gynecol 2002; 100 (1) 8-17
  • 3 Marshall LM, Spiegelman D, Barbieri RL , et al. Variation in the incidence of uterine leiomyoma among premenopausal women by age and race. Obstet Gynecol 1997; 90 (6) 967-973
  • 4 Cramer SF, Patel A. The frequency of uterine leiomyomas. Am J Clin Pathol 1990; 94 (4) 435-438
  • 5 Bulun SE. Uterine fibroids. N Engl J Med 2013; 369 (14) 1344-1355
  • 6 Baird DD, Dunson DB, Hill MC, Cousins D, Schectman JM. High cumulative incidence of uterine leiomyoma in black and white women: ultrasound evidence. Am J Obstet Gynecol 2003; 188 (1) 100-107
  • 7 Walker CL, Stewart EA. Uterine fibroids: the elephant in the room. Science 2005; 308 (5728) 1589-1592
  • 8 Peddada SD, Laughlin SK, Miner K , et al. Growth of uterine leiomyomata among premenopausal black and white women. Proc Natl Acad Sci U S A 2008; 105 (50) 19887-19892
  • 9 Townsend DE, Sparkes RS, Baluda MC, McClelland G. Unicellular histogenesis of uterine leiomyomas as determined by electrophoresis by glucose-6-phosphate dehydrogenase. Am J Obstet Gynecol 1970; 107 (8) 1168-1173
  • 10 Linder D, Gartler SM. Glucose-6-phosphate dehydrogenase mosaicism: utilization as a cell marker in the study of leiomyomas. Science 1965; 150 (3692) 67-69
  • 11 Tal R, Segars JH. The role of angiogenic factors in fibroid pathogenesis: potential implications for future therapy. Hum Reprod Update 2014; 20 (2) 194-216
  • 12 Mehine M, Kaasinen E, Mäkinen N , et al. Characterization of uterine leiomyomas by whole-genome sequencing. N Engl J Med 2013; 369 (1) 43-53
  • 13 Kim JJ, Kurita T, Bulun SE. Progesterone action in endometrial cancer, endometriosis, uterine fibroids, and breast cancer. Endocr Rev 2013; 34 (1) 130-162
  • 14 Flake GP, Moore AB, Sutton D , et al. The natural history of uterine leiomyomas: light and electron microscopic studies of fibroid phases, interstitial ischemia, inanosis, and reclamation. Obstet Gynecol Int 2013; 2013: 528376
  • 15 Cardozo ER, Clark AD, Banks NK, Henne MB, Stegmann BJ, Segars JH. The estimated annual cost of uterine leiomyomata in the United States. Am J Obstet Gynecol 2012; 206 (3) 211.e1-211.e9
  • 16 Freed MM, Spies JB. Uterine artery embolization for fibroids: a review of current outcomes. Semin Reprod Med 2010; 28 (3) 235-241
  • 17 Al Hilli MM, Stewart EA. Magnetic resonance-guided focused ultrasound surgery. Semin Reprod Med 2010; 28 (3) 242-249
  • 18 Donnez J, Tatarchuk TF, Bouchard P , et al; PEARL I Study Group. Ulipristal acetate versus placebo for fibroid treatment before surgery. N Engl J Med 2012; 366 (5) 409-420
  • 19 Donnez J, Tomaszewski J, Vázquez F , et al; PEARL II Study Group. Ulipristal acetate versus leuprolide acetate for uterine fibroids. N Engl J Med 2012; 366 (5) 421-432
  • 20 Sabry M, Al-Hendy A. Innovative oral treatments of uterine leiomyoma. Obstet Gynecol Int 2012; 2012: 943635
  • 21 Roshdy E, Rajaratnam V, Maitra S, Sabry M, Allah AS, Al-Hendy A. Treatment of symptomatic uterine fibroids with green tea extract: a pilot randomized controlled clinical study. Int J Womens Health 2013; 5: 477-486
  • 22 Halder SK, Osteen KG, Al-Hendy A. Vitamin D3 inhibits expression and activities of matrix metalloproteinase-2 and -9 in human uterine fibroid cells. Hum Reprod 2013; 28 (9) 2407-2416
  • 23 Halder SK, Sharan C, Al-Hendy A. 1,25-dihydroxyvitamin D3 treatment shrinks uterine leiomyoma tumors in the Eker rat model. Biol Reprod 2012; 86 (4) 116
  • 24 Ono M, Qiang W, Serna VA , et al. Role of stem cells in human uterine leiomyoma growth. PLoS ONE 2012; 7 (5) e36935
  • 25 Mas A, Cervelló I, Gil-Sanchis C , et al. Identification and characterization of the human leiomyoma side population as putative tumor-initiating cells. Fertil Steril 2012; 98 (3) 741-751.e6
  • 26 Ishikawa H, Fenkci V, Marsh EE , et al. CCAAT/enhancer binding protein beta regulates aromatase expression via multiple and novel cis-regulatory sequences in uterine leiomyoma. J Clin Endocrinol Metab 2008; 93 (3) 981-991
  • 27 Lamminen S, Rantala I, Helin H, Rorarius M, Tuimala R. Proliferative activity of human uterine leiomyoma cells as measured by automatic image analysis. Gynecol Obstet Invest 1992; 34 (2) 111-114
  • 28 Ishikawa H, Ishi K, Serna VA, Kakazu R, Bulun SE, Kurita T. Progesterone is essential for maintenance and growth of uterine leiomyoma. Endocrinology 2010; 151 (6) 2433-2442
  • 29 Hassan MH, Salama SA, Arafa HM, Hamada FM, Al-Hendy A. Adenovirus-mediated delivery of a dominant-negative estrogen receptor gene in uterine leiomyoma cells abrogates estrogen- and progesterone-regulated gene expression. J Clin Endocrinol Metab 2007; 92 (10) 3949-3957
  • 30 Cermik D, Arici A, Taylor HS. Coordinated regulation of HOX gene expression in myometrium and uterine leiomyoma. Fertil Steril 2002; 78 (5) 979-984
  • 31 Mangelsdorf DJ, Thummel C, Beato M , et al. The nuclear receptor superfamily: the second decade. Cell 1995; 83 (6) 835-839
  • 32 Robinson-Rechavi M, Escriva Garcia H, Laudet V. The nuclear receptor superfamily. J Cell Sci 2003; 116 (Pt 4) 585-586
  • 33 Lessey BA, Alexander PS, Horwitz KB. The subunit structure of human breast cancer progesterone receptors: characterization by chromatography and photoaffinity labeling. Endocrinology 1983; 112 (4) 1267-1274
  • 34 Kastner P, Krust A, Turcotte B , et al. Two distinct estrogen-regulated promoters generate transcripts encoding the two functionally different human progesterone receptor forms A and B. EMBO J 1990; 9 (5) 1603-1614
  • 35 Gronemeyer H, Meyer ME, Bocquel MT, Kastner P, Turcotte B, Chambon P. Progestin receptors: isoforms and antihormone action. J Steroid Biochem Mol Biol 1991; 40 (1-3) 271-278
  • 36 Vegeto E, Shahbaz MM, Wen DX, Goldman ME, O'Malley BW, McDonnell DP. Human progesterone receptor A form is a cell- and promoter-specific repressor of human progesterone receptor B function. Mol Endocrinol 1993; 7 (10) 1244-1255
  • 37 Tung L, Mohamed MK, Hoeffler JP, Takimoto GS, Horwitz KB. Antagonist-occupied human progesterone B-receptors activate transcription without binding to progesterone response elements and are dominantly inhibited by A-receptors. Mol Endocrinol 1993; 7 (10) 1256-1265
  • 38 McDonnell DP, Shahbaz MM, Vegeto E, Goldman ME. The human progesterone receptor A-form functions as a transcriptional modulator of mineralocorticoid receptor transcriptional activity. J Steroid Biochem Mol Biol 1994; 48 (5-6) 425-432
  • 39 Edwards DP. The role of coactivators and corepressors in the biology and mechanism of action of steroid hormone receptors. J Mammary Gland Biol Neoplasia 2000; 5 (3) 307-324
  • 40 Tetel MJ, Giangrande PH, Leonhardt SA, McDonnell DP, Edwards DP. Hormone-dependent interaction between the amino- and carboxyl-terminal domains of progesterone receptor in vitro and in vivo. Mol Endocrinol 1999; 13 (6) 910-924
  • 41 Tung L, Abdel-Hafiz H, Shen T , et al. Progesterone receptors (PR)-B and -A regulate transcription by different mechanisms: AF-3 exerts regulatory control over coactivator binding to PR-B. Mol Endocrinol 2006; 20 (11) 2656-2670
  • 42 Brandon DD, Bethea CL, Strawn EY , et al. Progesterone receptor messenger ribonucleic acid and protein are overexpressed in human uterine leiomyomas. Am J Obstet Gynecol 1993; 169 (1) 78-85
  • 43 Englund K, Blanck A, Gustavsson I , et al. Sex steroid receptors in human myometrium and fibroids: changes during the menstrual cycle and gonadotropin-releasing hormone treatment. J Clin Endocrinol Metab 1998; 83 (11) 4092-4096
  • 44 Nisolle M, Gillerot S, Casanas-Roux F, Squifflet J, Berliere M, Donnez J. Immunohistochemical study of the proliferation index, oestrogen receptors and progesterone receptors A and B in leiomyomata and normal myometrium during the menstrual cycle and under gonadotrophin-releasing hormone agonist therapy. Hum Reprod 1999; 14 (11) 2844-2850
  • 45 Ishikawa H, Reierstad S, Demura M , et al. High aromatase expression in uterine leiomyoma tissues of African-American women. J Clin Endocrinol Metab 2009; 94 (5) 1752-1756
  • 46 Mulac-Jericevic B, Lydon JP, DeMayo FJ, Conneely OM. Defective mammary gland morphogenesis in mice lacking the progesterone receptor B isoform. Proc Natl Acad Sci U S A 2003; 100 (17) 9744-9749
  • 47 Mulac-Jericevic B, Mullinax RA, DeMayo FJ, Lydon JP, Conneely OM. Subgroup of reproductive functions of progesterone mediated by progesterone receptor-B isoform. Science 2000; 289 (5485) 1751-1754
  • 48 Kawaguchi K, Fujii S, Konishi I, Nanbu Y, Nonogaki H, Mori T. Mitotic activity in uterine leiomyomas during the menstrual cycle. Am J Obstet Gynecol 1989; 160 (3) 637-641
  • 49 Murphy AA, Kettel LM, Morales AJ, Roberts VJ, Yen SS. Regression of uterine leiomyomata in response to the antiprogesterone RU 486. J Clin Endocrinol Metab 1993; 76 (2) 513-517
  • 50 Chwalisz K, Perez MC, Demanno D, Winkel C, Schubert G, Elger W. Selective progesterone receptor modulator development and use in the treatment of leiomyomata and endometriosis. Endocr Rev 2005; 26 (3) 423-438
  • 51 Spitz IM. Clinical utility of progesterone receptor modulators and their effect on the endometrium. Curr Opin Obstet Gynecol 2009; 21 (4) 318-324
  • 52 Chabbert-Buffet N, Meduri G, Bouchard P, Spitz IM. Selective progesterone receptor modulators and progesterone antagonists: mechanisms of action and clinical applications. Hum Reprod Update 2005; 11 (3) 293-307
  • 53 Islam MS, Protic O, Giannubilo SR , et al. Uterine leiomyoma: available medical treatments and new possible therapeutic options. J Clin Endocrinol Metab 2013; 98 (3) 921-934
  • 54 Talaulikar VS, Manyonda IT. Ulipristal acetate: a novel option for the medical management of symptomatic uterine fibroids. Adv Ther 2012; 29 (8) 655-663
  • 55 Bouchard P, Chabbert-Buffet N, Fauser BC. Selective progesterone receptor modulators in reproductive medicine: pharmacology, clinical efficacy and safety. Fertil Steril 2011; 96 (5) 1175-1189
  • 56 Yin P, Lin Z, Reierstad S , et al. Transcription factor KLF11 integrates progesterone receptor signaling and proliferation in uterine leiomyoma cells. Cancer Res 2010; 70 (4) 1722-1730
  • 57 Yin P, Roqueiro D, Huang L , et al. Genome-wide progesterone receptor binding: cell type-specific and shared mechanisms in T47D breast cancer cells and primary leiomyoma cells. PLoS ONE 2012; 7 (1) e29021
  • 58 Chen W, Ohara N, Wang J , et al. A novel selective progesterone receptor modulator asoprisnil (J867) inhibits proliferation and induces apoptosis in cultured human uterine leiomyoma cells in the absence of comparable effects on myometrial cells. J Clin Endocrinol Metab 2006; 91 (4) 1296-1304
  • 59 Luo X, Yin P, Coon V JS, Cheng YH, Wiehle RD, Bulun SE. The selective progesterone receptor modulator CDB4124 inhibits proliferation and induces apoptosis in uterine leiomyoma cells. Fertil Steril 2010; 93 (8) 2668-2673
  • 60 Maruo T, Ohara N, Matsuo H , et al. Effects of levonorgestrel-releasing IUS and progesterone receptor modulator PRM CDB-2914 on uterine leiomyomas. Contraception 2007; 75 (6, Suppl): S99-S103
  • 61 Xu Q, Takekida S, Ohara N , et al. Progesterone receptor modulator CDB-2914 down-regulates proliferative cell nuclear antigen and Bcl-2 protein expression and up-regulates caspase-3 and poly(adenosine 5′-diphosphate-ribose) polymerase expression in cultured human uterine leiomyoma cells. J Clin Endocrinol Metab 2005; 90 (2) 953-961
  • 62 Wang J, Ohara N, Wang Z , et al. A novel selective progesterone receptor modulator asoprisnil (J867) down-regulates the expression of EGF, IGF-I, TGFbeta3 and their receptors in cultured uterine leiomyoma cells. Hum Reprod 2006; 21 (7) 1869-1877
  • 63 Xu Q, Ohara N, Chen W , et al. Progesterone receptor modulator CDB-2914 down-regulates vascular endothelial growth factor, adrenomedullin and their receptors and modulates progesterone receptor content in cultured human uterine leiomyoma cells. Hum Reprod 2006; 21 (9) 2408-2416
  • 64 Yoshida S, Ohara N, Xu Q , et al. Cell-type specific actions of progesterone receptor modulators in the regulation of uterine leiomyoma growth. Semin Reprod Med 2010; 28 (3) 260-273
  • 65 Xu Q, Ohara N, Liu J , et al. Selective progesterone receptor modulator asoprisnil induces endoplasmic reticulum stress in cultured human uterine leiomyoma cells. Am J Physiol Endocrinol Metab 2007; 293 (4) E1002-E1011
  • 66 Sasaki H, Ohara N, Xu Q , et al. A novel selective progesterone receptor modulator asoprisnil activates tumor necrosis factor-related apoptosis-inducing ligand (TRAIL)-mediated signaling pathway in cultured human uterine leiomyoma cells in the absence of comparable effects on myometrial cells. J Clin Endocrinol Metab 2007; 92 (2) 616-623
  • 67 Morikawa A, Ohara N, Xu Q , et al. Selective progesterone receptor modulator asoprisnil down-regulates collagen synthesis in cultured human uterine leiomyoma cells through up-regulating extracellular matrix metalloproteinase inducer. Hum Reprod 2008; 23 (4) 944-951
  • 68 Xu Q, Ohara N, Liu J , et al. Progesterone receptor modulator CDB-2914 induces extracellular matrix metalloproteinase inducer in cultured human uterine leiomyoma cells. Mol Hum Reprod 2008; 14 (3) 181-191
  • 69 Weissman IL. Stem cells: units of development, units of regeneration, and units in evolution. Cell 2000; 100 (1) 157-168
  • 70 Li L, Clevers H. Coexistence of quiescent and active adult stem cells in mammals. Science 2010; 327 (5965) 542-545
  • 71 Jordan CT, Guzman ML, Noble M. Cancer stem cells. N Engl J Med 2006; 355 (12) 1253-1261
  • 72 Schofield R. The relationship between the spleen colony-forming cell and the haemopoietic stem cell. Blood Cells 1978; 4 (1-2) 7-25
  • 73 Ono M, Maruyama T, Masuda H , et al. Side population in human uterine myometrium displays phenotypic and functional characteristics of myometrial stem cells. Proc Natl Acad Sci U S A 2007; 104 (47) 18700-18705
  • 74 Szotek PP, Chang HL, Zhang L , et al. Adult mouse myometrial label-retaining cells divide in response to gonadotropin stimulation. Stem Cells 2007; 25 (5) 1317-1325
  • 75 Arango NA, Szotek PP, Manganaro TF, Oliva E, Donahoe PK, Teixeira J. Conditional deletion of beta-catenin in the mesenchyme of the developing mouse uterus results in a switch to adipogenesis in the myometrium. Dev Biol 2005; 288 (1) 276-283
  • 76 Mäkinen N, Mehine M, Tolvanen J , et al. MED12, the mediator complex subunit 12 gene, is mutated at high frequency in uterine leiomyomas. Science 2011; 334 (6053) 252-255
  • 77 McGuire MM, Yatsenko A, Hoffner L, Jones M, Surti U, Rajkovic A. Whole exome sequencing in a random sample of North American women with leiomyomas identifies MED12 mutations in majority of uterine leiomyomas. PLoS ONE 2012; 7 (3) e33251
  • 78 Bertsch E, Qiang W, Zhang Q , et al. MED12 and HMGA2 mutations: two independent genetic events in uterine leiomyoma and leiomyosarcoma. Mod Pathol 2014; 27 (8) 1144-1153
  • 79 Maruyama T, Miyazaki K, Masuda H, Ono M, Uchida H, Yoshimura Y. Review: human uterine stem/progenitor cells: implications for uterine physiology and pathology. Placenta 2013; 34 (Suppl): S68-S72
  • 80 Zhou S, Yi T, Shen K, Zhang B, Huang F, Zhao X. Hypoxia: the driving force of uterine myometrial stem cells differentiation into leiomyoma cells. Med Hypotheses 2011; 77 (6) 985-986
  • 81 Challen GA, Little MH. A side order of stem cells: the SP phenotype. Stem Cells 2006; 24 (1) 3-12
  • 82 Gargett CE, Masuda H. Adult stem cells in the endometrium. Mol Hum Reprod 2010; 16 (11) 818-834
  • 83 Goodell MA, Brose K, Paradis G, Conner AS, Mulligan RC. Isolation and functional properties of murine hematopoietic stem cells that are replicating in vivo. J Exp Med 1996; 183 (4) 1797-1806
  • 84 Golebiewska A, Brons NH, Bjerkvig R, Niclou SP. Critical appraisal of the side population assay in stem cell and cancer stem cell research. Cell Stem Cell 2011; 8 (2) 136-147
  • 85 Yin P, Ono M, Moravek MB , et al. Human uterine leiomyoma stem/progenitor cells expressing CD34 and CD49b initiate tumors in vivo. J Clin Endocrinol Metab 2015; 100 (4) E601-E606
  • 86 Chang HL, Senaratne TN, Zhang L , et al. Uterine leiomyomas exhibit fewer stem/progenitor cell characteristics when compared with corresponding normal myometrium. Reprod Sci 2010; 17 (2) 158-167
  • 87 Ono M, Yin P, Navarro A , et al. Paracrine activation of WNT/β-catenin pathway in uterine leiomyoma stem cells promotes tumor growth. Proc Natl Acad Sci U S A 2013; 110 (42) 17053-17058
  • 88 Gottardi CJ, Königshoff M. Considerations for targeting β-catenin signaling in fibrosis. Am J Respir Crit Care Med 2013; 187 (6) 566-568
  • 89 Lam AP, Gottardi CJ. β-catenin signaling: a novel mediator of fibrosis and potential therapeutic target. Curr Opin Rheumatol 2011; 23 (6) 562-567
  • 90 Tanwar PS, Lee HJ, Zhang L , et al. Constitutive activation of Beta-catenin in uterine stroma and smooth muscle leads to the development of mesenchymal tumors in mice. Biol Reprod 2009; 81 (3) 545-552
  • 91 Kim S, Xu X, Hecht A, Boyer TG. Mediator is a transducer of Wnt/beta-catenin signaling. J Biol Chem 2006; 281 (20) 14066-14075
  • 92 Fillmore CM, Gupta PB, Rudnick JA , et al. Estrogen expands breast cancer stem-like cells through paracrine FGF/Tbx3 signaling. Proc Natl Acad Sci U S A 2010; 107 (50) 21737-21742
  • 93 Roarty K, Rosen JM. Wnt and mammary stem cells: hormones cannot fly wingless. Curr Opin Pharmacol 2010; 10 (6) 643-649
  • 94 Asselin-Labat ML, Vaillant F, Sheridan JM , et al. Control of mammary stem cell function by steroid hormone signalling. Nature 2010; 465 (7299) 798-802
  • 95 Vares G, Cui X, Wang B, Nakajima T, Nenoi M. Generation of breast cancer stem cells by steroid hormones in irradiated human mammary cell lines. PLoS ONE 2013; 8 (10) e77124
  • 96 Cittelly DM, Finlay-Schultz J, Howe EN , et al. Progestin suppression of miR-29 potentiates dedifferentiation of breast cancer cells via KLF4. Oncogene 2013; 32 (20) 2555-2564
  • 97 Montales MT, Rahal OM, Kang J , et al. Repression of mammosphere formation of human breast cancer cells by soy isoflavone genistein and blueberry polyphenolic acids suggests diet-mediated targeting of cancer stem-like/progenitor cells. Carcinogenesis 2012; 33 (3) 652-660
  • 98 Qiang W, Liu Z, Serna VA , et al. Down-regulation of miR-29b is essential for pathogenesis of uterine leiomyoma. Endocrinology 2014; 155 (3) 663-669