Skip to main content
Top
Published in: BMC Women's Health 1/2019

Open Access 01-12-2019 | Progesterone | Research article

Myometrial progesterone hyper-responsiveness associated with increased risk of human uterine fibroids

Authors: Mona Omar, Archana Laknaur, Ayman Al-Hendy, Qiwei Yang

Published in: BMC Women's Health | Issue 1/2019

Login to get access

Abstract

Background

Uterine Fibroids (UFs) growth is ovarian steroid-dependent. Previous studies have shown that estrogen and progesterone play an important role in UF development. However, the mechanism underlying progesterone induced UF pathogenesis is largely unknown. In this study, we determined the expression of progesterone receptor and compared the expression level of progesterone-regulated genes (PRGs) in human myometrial cells from normal uteri (MyoN) versus uteri with UFs (MyoF) in response to progesterone.

Methods

Primary human myometrial cells were isolated from premenopausal patients with structurally normal uteri (PrMyoN). Primary human myometrial cells were also isolated from uterus with UFs (PrMyoF). Isolated tissues were excised at least 2 cm from the closest UFs lesion(s). Progesterone receptor (PR) expression was assessed using Western blot (WB). Expression levels of 15 PRGs were measured by qRT-PCR in PrMyoN and PrMyoF cells in the presence or absence of progesterone.

Results

WB analysis revealed higher expression levels of PR in PrMyoF cells as compared to PrMyoN cells. Furthermore, we compared the expression patterns of 15 UF-related PRGs in PrMyoN and PrMyoF primary cells in response to progesterone hormone treatment. Our studies demonstrated that five PRGs including Bcl2, FOXO1A, SCGB2A2, CYP26a1 and MMP11 exhibited significant progesterone-hyper-responsiveness in human PrMyoF cells as compared to PrMyoN cells (P < 0.05). Another seven PRGs, including CIDEC, CANP6, ADHL5, ALDHA1, MT1E, KIK6, HHI showed gain in repression in response to progesterone treatment (P > 0.05). Importantly, these genes play crucial roles in cell proliferation, apoptosis, cell cycle, tissue remodeling and tumorigenesis in the development of UFs.

Conclusion

These data support the idea that progesterone acts as contributing mechanism in the origin of UFs. Identification and analysis of these PRGs will help to further understand the role of progesterone in UF development.
Literature
1.
go back to reference Cramer SF, Patel A. The frequency of uterine leiomyomas. Am J Clin Pathol. 1990;94:435–8.CrossRef Cramer SF, Patel A. The frequency of uterine leiomyomas. Am J Clin Pathol. 1990;94:435–8.CrossRef
2.
go back to reference Julie Kim J, Sefton EC. The role of progesterone signaling in the pathogenesis of uterine leiomyoma. Mol Cell Endocrinol. 2012;358:223–31.CrossRef Julie Kim J, Sefton EC. The role of progesterone signaling in the pathogenesis of uterine leiomyoma. Mol Cell Endocrinol. 2012;358:223–31.CrossRef
3.
go back to reference Cermik D, Arici A, Taylor HS. Coordinated regulation of HOX gene expression in myometrium and uterine leiomyoma. Fertil Steril. 2002;78:979–84.CrossRef Cermik D, Arici A, Taylor HS. Coordinated regulation of HOX gene expression in myometrium and uterine leiomyoma. Fertil Steril. 2002;78:979–84.CrossRef
4.
5.
go back to reference Ishikawa H, Ishi K, Serna VA, Kakazu R, Bulun SE, Kurita T. Progesterone is essential for maintenance and growth of uterine leiomyoma. Endocrinology. 2010;151:2433–42.CrossRef Ishikawa H, Ishi K, Serna VA, Kakazu R, Bulun SE, Kurita T. Progesterone is essential for maintenance and growth of uterine leiomyoma. Endocrinology. 2010;151:2433–42.CrossRef
6.
go back to reference Carr BR, Marshburn PB, Weatherall PT, Bradshaw KD, Breslau NA, Byrd W, Roark M, Steinkampf MP. An evaluation of the effect of gonadotropinreleasing hormone analogs and medroxyprogesterone acetate on uterine leiomyomata volume by magnetic resonance imaging: a prospective, randomized, double blind, placebo controlled, crossover trial. J Clin Endocrinol Metab. 1993;76:1217–23.PubMed Carr BR, Marshburn PB, Weatherall PT, Bradshaw KD, Breslau NA, Byrd W, Roark M, Steinkampf MP. An evaluation of the effect of gonadotropinreleasing hormone analogs and medroxyprogesterone acetate on uterine leiomyomata volume by magnetic resonance imaging: a prospective, randomized, double blind, placebo controlled, crossover trial. J Clin Endocrinol Metab. 1993;76:1217–23.PubMed
7.
go back to reference Friedman AJ, Daly M, Juneau-Norcross M, Rein MS, Fine C, Gleason R, Leboff M. A prospective, randomized trial of gonadotropin-releasing hormone agonist plus estrogen-progestin or progestin “add-back” regimens for women with leiomyomata uteri. J Clin Endocrinol Metab. 1993;76:1439–45.PubMed Friedman AJ, Daly M, Juneau-Norcross M, Rein MS, Fine C, Gleason R, Leboff M. A prospective, randomized trial of gonadotropin-releasing hormone agonist plus estrogen-progestin or progestin “add-back” regimens for women with leiomyomata uteri. J Clin Endocrinol Metab. 1993;76:1439–45.PubMed
8.
go back to reference Friedman AJ, Daly M, Juneau-Norcross M, Gleason R, Rein MS, LeBoff M. Long-term medical therapy for leiomyomata uteri: a prospective, randomized study of leuprolide acetate depot plus either oestrogen-progestin or progestin ‘add-back’ for 2 years. Hum Reprod. 1994;9:1618–25.CrossRef Friedman AJ, Daly M, Juneau-Norcross M, Gleason R, Rein MS, LeBoff M. Long-term medical therapy for leiomyomata uteri: a prospective, randomized study of leuprolide acetate depot plus either oestrogen-progestin or progestin ‘add-back’ for 2 years. Hum Reprod. 1994;9:1618–25.CrossRef
9.
go back to reference Bagaria M, Suneja A, Vaid NB, Guleria K, Mishra K. Low-dose mifepristone in treatment of uterine leiomyoma: a randomised double-blind placebo-controlled clinical trial. Aust N Z J Obstet Gynecol. 2009;49:77–83.CrossRef Bagaria M, Suneja A, Vaid NB, Guleria K, Mishra K. Low-dose mifepristone in treatment of uterine leiomyoma: a randomised double-blind placebo-controlled clinical trial. Aust N Z J Obstet Gynecol. 2009;49:77–83.CrossRef
10.
go back to reference Engman M, Granberg S, Williams AR, Meng CX, Lalitkumar PG, Gemzell- Danielsson K. Mifepristone for treatment of uterine leiomyoma. A prospective randomized placebo controlled trial. Hum Reprod. 2009;24:1870–9.CrossRef Engman M, Granberg S, Williams AR, Meng CX, Lalitkumar PG, Gemzell- Danielsson K. Mifepristone for treatment of uterine leiomyoma. A prospective randomized placebo controlled trial. Hum Reprod. 2009;24:1870–9.CrossRef
11.
go back to reference Spitz IM. Clinical utility of progesterone receptor modulators and their effect on the endometrium. Curr Opin Obstet Gynecol. 2009;21:318–24.CrossRef Spitz IM. Clinical utility of progesterone receptor modulators and their effect on the endometrium. Curr Opin Obstet Gynecol. 2009;21:318–24.CrossRef
12.
go back to reference Misrahi M, Atger M, d’Auriol L, Loosfelt H, Meriel C, Fridlansky F, Guiochon-Mantel A, Galibert F, Milgrom E. Complete amino acid sequence of the human progesterone receptor deduced from cloned cDNA. Biochem Biophys Res Commun. 1987;143:740–8.CrossRef Misrahi M, Atger M, d’Auriol L, Loosfelt H, Meriel C, Fridlansky F, Guiochon-Mantel A, Galibert F, Milgrom E. Complete amino acid sequence of the human progesterone receptor deduced from cloned cDNA. Biochem Biophys Res Commun. 1987;143:740–8.CrossRef
13.
go back to reference Mangelsdorf DJ, Thummel C, Beato M, Herrlich P, Schutz G, Umesono K, Blumberg B, Kastner P, Mark M, Chambon P, Evans RM. The nuclear receptor superfamily: the second decade. Cell. 1995;83:835–9.CrossRef Mangelsdorf DJ, Thummel C, Beato M, Herrlich P, Schutz G, Umesono K, Blumberg B, Kastner P, Mark M, Chambon P, Evans RM. The nuclear receptor superfamily: the second decade. Cell. 1995;83:835–9.CrossRef
14.
go back to reference Robinson-Rechavi M, Escriva Garcia H, Laudet V. The nuclear receptor superfamily. J Cell Sci. 2003;116:585–6.CrossRef Robinson-Rechavi M, Escriva Garcia H, Laudet V. The nuclear receptor superfamily. J Cell Sci. 2003;116:585–6.CrossRef
15.
go back to reference Kastner P, Krust A, Turcotte B, Stropp U, Tora L, Gronemeyer H, Chambon P. Two distinct estrogen-regulated promoters generate transcripts encoding the two functionally different human progesterone receptor forms a and B. EMBO J. 1990;9:1603–14.CrossRef Kastner P, Krust A, Turcotte B, Stropp U, Tora L, Gronemeyer H, Chambon P. Two distinct estrogen-regulated promoters generate transcripts encoding the two functionally different human progesterone receptor forms a and B. EMBO J. 1990;9:1603–14.CrossRef
16.
go back to reference Samalecos A, Gellersen B. Systematic expression analysis and antibody screening do not support the existence of naturally occurring progesterone receptor (PR)-C, PR-M, or other truncated PR isoforms. Endocrinology. 2008;149:5872–87.CrossRef Samalecos A, Gellersen B. Systematic expression analysis and antibody screening do not support the existence of naturally occurring progesterone receptor (PR)-C, PR-M, or other truncated PR isoforms. Endocrinology. 2008;149:5872–87.CrossRef
17.
go back to reference Gellersen B, Fernandes MS, Brosens JJ. Non-genomic progesterone actions in female reproduction. Hum Reprod Update. 2009;15:119–38.CrossRef Gellersen B, Fernandes MS, Brosens JJ. Non-genomic progesterone actions in female reproduction. Hum Reprod Update. 2009;15:119–38.CrossRef
18.
go back to reference Cook JD, Davis BJ, Cai SL, Barrett JC, Conti CJ, Walker CL. Interaction between genetic susceptibility and early-life environmental exposure determines tumor-suppressor-gene penetrance. Proc Natl Acad Sci U S A. 2005;102(24):8644–9.CrossRef Cook JD, Davis BJ, Cai SL, Barrett JC, Conti CJ, Walker CL. Interaction between genetic susceptibility and early-life environmental exposure determines tumor-suppressor-gene penetrance. Proc Natl Acad Sci U S A. 2005;102(24):8644–9.CrossRef
20.
go back to reference Mas A, Stone L, O'Connor PM, Yang Q, Kleven D, Simon C, Walker CL, Al-Hendy A. Developmental exposure to endocrine disruptors expands murine myometrial stem cell compartment as a prerequisite to leiomyoma tumorigenesis. Stem Cells. 2017;35(3):666–78. https://doi.org/10.1002/stem.2519 Epub 2016 Nov 11.CrossRefPubMed Mas A, Stone L, O'Connor PM, Yang Q, Kleven D, Simon C, Walker CL, Al-Hendy A. Developmental exposure to endocrine disruptors expands murine myometrial stem cell compartment as a prerequisite to leiomyoma tumorigenesis. Stem Cells. 2017;35(3):666–78. https://​doi.​org/​10.​1002/​stem.​2519 Epub 2016 Nov 11.CrossRefPubMed
21.
go back to reference Lauren E, Fernung P, Kimya Jones Y, Aymara Mas Z, Daniel Kleven Y, Jennifer L, Waller X, Al-Hendy A. Expanding upon the Human Myometrial Stem Cell Hypothesis and the Role of Race, Hormones., Age, and Parity in a Profibroid Environment. Am J Pathol. 2018;188:10. Lauren E, Fernung P, Kimya Jones Y, Aymara Mas Z, Daniel Kleven Y, Jennifer L, Waller X, Al-Hendy A. Expanding upon the Human Myometrial Stem Cell Hypothesis and the Role of Race, Hormones., Age, and Parity in a Profibroid Environment. Am J Pathol. 2018;188:10.
22.
go back to reference 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.CrossRef 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.CrossRef
25.
go back to reference Pfaffl MW, Horgan GW, Dempfle L. Relative expression software tool (REST) for group-wise comparison and statistical analysis of relative expression results in real-time PCR. Nucleic Acids Res. 2002;30:e36.CrossRef Pfaffl MW, Horgan GW, Dempfle L. Relative expression software tool (REST) for group-wise comparison and statistical analysis of relative expression results in real-time PCR. Nucleic Acids Res. 2002;30:e36.CrossRef
26.
go back to reference Jeong JW, et al. Identification of murine uterine genes regulated in a ligand-dependent manner by the progesterone receptor. Genes Endocrinology, August. 2005;146(8):3490–505. Jeong JW, et al. Identification of murine uterine genes regulated in a ligand-dependent manner by the progesterone receptor. Genes Endocrinology, August. 2005;146(8):3490–505.
27.
go back to reference shimomura Y, et al. Up-regulation by progesterone of proliferating cell nuclear antigen and epidermal growth factor expression in human uterine leiomyoma. J Clin Endocrinol Metab. 1998;83:6. shimomura Y, et al. Up-regulation by progesterone of proliferating cell nuclear antigen and epidermal growth factor expression in human uterine leiomyoma. J Clin Endocrinol Metab. 1998;83:6.
28.
go back to reference Burney et al. Progesterone resistance in endometriosis endocrinology, August 2007, 148(8):3814–3826. Burney et al. Progesterone resistance in endometriosis endocrinology, August 2007, 148(8):3814–3826.
29.
go back to reference 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;169:78–85.CrossRef 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;169:78–85.CrossRef
30.
go back to reference Kerr JER, Wylie AH, Currie AR. Apoptosis: a basic biological phenomenon with wide-ranging implications in tissue kinetics. Br J Cancer. 1972;26:239–57.CrossRef Kerr JER, Wylie AH, Currie AR. Apoptosis: a basic biological phenomenon with wide-ranging implications in tissue kinetics. Br J Cancer. 1972;26:239–57.CrossRef
31.
go back to reference Korsmyer SJ. Bcl-2 initiates a new categoly of oncogenes: regulators of cell death. Blood. 1992;80:879–86. Korsmyer SJ. Bcl-2 initiates a new categoly of oncogenes: regulators of cell death. Blood. 1992;80:879–86.
32.
go back to reference Mcdonnell TJ, Deane N, Platt FM, et al. BCL-2 immunoglobulin transgenic mice demonstrate extended B cell survival and follicular lymphoproliferation. Cell. 1989;57:79–88.CrossRef Mcdonnell TJ, Deane N, Platt FM, et al. BCL-2 immunoglobulin transgenic mice demonstrate extended B cell survival and follicular lymphoproliferation. Cell. 1989;57:79–88.CrossRef
33.
go back to reference Matsuo M, Maruo T, Samoto T. Increased expression of Bcl-2 protein in human uterine leiomyoma and its up-regulation by progesterone. J Clin Endocrinol Metab. 1997;82:293–9.PubMed Matsuo M, Maruo T, Samoto T. Increased expression of Bcl-2 protein in human uterine leiomyoma and its up-regulation by progesterone. J Clin Endocrinol Metab. 1997;82:293–9.PubMed
34.
go back to reference Christian M, Zhang X, Schneider-Merck T, Unterman TG, Gellersen B, White JO, Brosens JJ. Cyclic AMPinduced forkhead transcription factor, FKHR, cooperates with CCAAT/enhancer-binding protein in differentiating human endometrial stromal cells. J Biol Chem. 2002;277:20825–32.CrossRef Christian M, Zhang X, Schneider-Merck T, Unterman TG, Gellersen B, White JO, Brosens JJ. Cyclic AMPinduced forkhead transcription factor, FKHR, cooperates with CCAAT/enhancer-binding protein in differentiating human endometrial stromal cells. J Biol Chem. 2002;277:20825–32.CrossRef
35.
go back to reference Medema RH, Kops GJ, Bos JL, Burgering BM. AFX-like Forkhead transcription factors mediate cell-cyclecycle regulation by Ras and PKB through p27kip1. Nature. 2000;404:782–7.CrossRef Medema RH, Kops GJ, Bos JL, Burgering BM. AFX-like Forkhead transcription factors mediate cell-cyclecycle regulation by Ras and PKB through p27kip1. Nature. 2000;404:782–7.CrossRef
36.
go back to reference Labied et al. • Progestins regulate the expression and activity of the Forkhead transcription factor FOXO1 inmDifferentiating human endometrium. Mol Endocrinol, January. 2006;20(1):35–44.CrossRef Labied et al. • Progestins regulate the expression and activity of the Forkhead transcription factor FOXO1 inmDifferentiating human endometrium. Mol Endocrinol, January. 2006;20(1):35–44.CrossRef
37.
go back to reference Watson MA, Fleming TP. Isolation of differentially expressed sequence tags from human breast cancer. Cancer Res. 1994;54:4598–602.PubMed Watson MA, Fleming TP. Isolation of differentially expressed sequence tags from human breast cancer. Cancer Res. 1994;54:4598–602.PubMed
38.
go back to reference Fischer K, von Brünneck AC, Hornung D, et al. Differential expression of secretoglobins in normal ovary and in ovarian carcinoma--overexpression of mammaglobin-1 is linked to tumor progression. Arch Biochem Biophys. 2014;547:27–36.CrossRef Fischer K, von Brünneck AC, Hornung D, et al. Differential expression of secretoglobins in normal ovary and in ovarian carcinoma--overexpression of mammaglobin-1 is linked to tumor progression. Arch Biochem Biophys. 2014;547:27–36.CrossRef
39.
go back to reference Gong J, Sun Z, Li P. CIDE proteins and metabolic disorders. Curr Opin Lipidol. 2009;20(2):121–6.CrossRef Gong J, Sun Z, Li P. CIDE proteins and metabolic disorders. Curr Opin Lipidol. 2009;20(2):121–6.CrossRef
40.
go back to reference Yonezawa T, Kurata R, Kimura M, et al. Which CIDE are you on? Apoptosis and energy metabolism. Mol BioSyst. 2011;7(1):91–100.CrossRef Yonezawa T, Kurata R, Kimura M, et al. Which CIDE are you on? Apoptosis and energy metabolism. Mol BioSyst. 2011;7(1):91–100.CrossRef
41.
go back to reference Zhou Z, Yon Toh S, Chen Z, et al. Cidea-deficient mice have lean phenotype and are resistant to obesity. Nat Genet. 2003;35(1):49–56.CrossRef Zhou Z, Yon Toh S, Chen Z, et al. Cidea-deficient mice have lean phenotype and are resistant to obesity. Nat Genet. 2003;35(1):49–56.CrossRef
42.
go back to reference Inohara N, Koseki T, Chen S, Wu X, Nunez G. CIDE, a novel family of cell death activators with homology to the 45 kDa subunit of the DNA fragmentation factor. EMBO J. 1998;17:2526–33.CrossRef Inohara N, Koseki T, Chen S, Wu X, Nunez G. CIDE, a novel family of cell death activators with homology to the 45 kDa subunit of the DNA fragmentation factor. EMBO J. 1998;17:2526–33.CrossRef
43.
go back to reference Joseph et al. Myometrial cells undergo fibrotic transformation under the influence of transforming growth factor b-3 TGF-b3 . Fertility and Sterility_2009 march; Vol. 93, No. 5. Joseph et al. Myometrial cells undergo fibrotic transformation under the influence of transforming growth factor b-3 TGF-b3 . Fertility and Sterility_2009 march; Vol. 93, No. 5.
44.
go back to reference Burgess HA, Reiner O. Cleavage of doublecortin-like kinase by calpain releases an active kinase fragment from a microtubule anchorage domain. J Biol Chem. 2001;276:36397–403.CrossRef Burgess HA, Reiner O. Cleavage of doublecortin-like kinase by calpain releases an active kinase fragment from a microtubule anchorage domain. J Biol Chem. 2001;276:36397–403.CrossRef
45.
go back to reference Carafoli E, Molinari M. Calpain: a protease in search of a function? Biochem Biophys Res Commun. 1998;247:193–203.CrossRef Carafoli E, Molinari M. Calpain: a protease in search of a function? Biochem Biophys Res Commun. 1998;247:193–203.CrossRef
46.
go back to reference Ponnamperuma RM, Kirchhof SM, Trifiletti L, De Luca LM. Ovariectomy increases squamous metaplasia of the uterine horns and survival of SENCAR mice fed a vitamin A-deficient diet. Am J Clin Nutr. 1999;70:502–8.CrossRef Ponnamperuma RM, Kirchhof SM, Trifiletti L, De Luca LM. Ovariectomy increases squamous metaplasia of the uterine horns and survival of SENCAR mice fed a vitamin A-deficient diet. Am J Clin Nutr. 1999;70:502–8.CrossRef
47.
go back to reference Murphy G, Nagase H. Progress in matrix metalloproteinase research. Mol Asp Med. 2008;29:290–308.CrossRef Murphy G, Nagase H. Progress in matrix metalloproteinase research. Mol Asp Med. 2008;29:290–308.CrossRef
48.
go back to reference Nagase H, Visse R, Murphy G. Structure and function of matrix metalloproteinases and TIMPs. Cardiovasc Res. 2006;69:562–73.CrossRef Nagase H, Visse R, Murphy G. Structure and function of matrix metalloproteinases and TIMPs. Cardiovasc Res. 2006;69:562–73.CrossRef
49.
go back to reference Pei D, Majmudar G, Weiss SJ. Hydrolytic inactivation of a breast carcinoma cell–derived serpin by human stromelysin-3. J Biol Chem. 1994;269:25849–55.PubMed Pei D, Majmudar G, Weiss SJ. Hydrolytic inactivation of a breast carcinoma cell–derived serpin by human stromelysin-3. J Biol Chem. 1994;269:25849–55.PubMed
50.
go back to reference Matziari M, Dive V, Yiotakis A. Matrix metalloproteinase 11 (MMP- 11; stromelysin-3) and synthetic inhibitors. Med Res Rev. 2007;27:528–52.CrossRef Matziari M, Dive V, Yiotakis A. Matrix metalloproteinase 11 (MMP- 11; stromelysin-3) and synthetic inhibitors. Med Res Rev. 2007;27:528–52.CrossRef
51.
go back to reference Yousef GM, Diamandis EP. An overview of the kallikrein gene families in humans and other species: emerging candidate tumour markerssmall star, filled. Clin Biochem. 2003;36:443–52.CrossRef Yousef GM, Diamandis EP. An overview of the kallikrein gene families in humans and other species: emerging candidate tumour markerssmall star, filled. Clin Biochem. 2003;36:443–52.CrossRef
52.
go back to reference Yousef GM, Diamandis EP. Expanded human tissue kallikrein family–a novel panel of cancer biomarkers. Tumour Biol. 2002;23:185–92.CrossRef Yousef GM, Diamandis EP. Expanded human tissue kallikrein family–a novel panel of cancer biomarkers. Tumour Biol. 2002;23:185–92.CrossRef
53.
go back to reference Yousef GM, Diamandis EP. Human tissue kallikreins: a new enzymatic cascade pathway? Biol Chem. 2002;383:1045–57.CrossRef Yousef GM, Diamandis EP. Human tissue kallikreins: a new enzymatic cascade pathway? Biol Chem. 2002;383:1045–57.CrossRef
54.
go back to reference Bhoola KD, Figueroa CD, Worthy K. Bioregulation of kinins: kallikreins, kininogens, and kininases. Pharmacol Rev. 1992;44:1–80.PubMed Bhoola KD, Figueroa CD, Worthy K. Bioregulation of kinins: kallikreins, kininogens, and kininases. Pharmacol Rev. 1992;44:1–80.PubMed
55.
go back to reference Takamoto N, Zhao B, Tsai SY, DeMayo FJ. Identification of Indian hedgehog as a progesterone-responsive gene in the murine uterus. Mol Endocrinol. 2002;16:2338–48.CrossRef Takamoto N, Zhao B, Tsai SY, DeMayo FJ. Identification of Indian hedgehog as a progesterone-responsive gene in the murine uterus. Mol Endocrinol. 2002;16:2338–48.CrossRef
56.
go back to reference Matsumoto H, Zhao X, Das SK, Hogan BL, Dey SK. Indian hedgehog as a progesterone-responsive factor mediating epithelial-mesenchymal interactions in the mouse uterus. Dev Biol. 2002;245:280–90.CrossRef Matsumoto H, Zhao X, Das SK, Hogan BL, Dey SK. Indian hedgehog as a progesterone-responsive factor mediating epithelial-mesenchymal interactions in the mouse uterus. Dev Biol. 2002;245:280–90.CrossRef
57.
go back to reference Talbi S, Hamilton AE, Vo KC, Tulac S, Overgaard MT, Dosiou C, Le Shay N, Nezhat CN, Kempson R, Lessey BA, Nayak NR, Giudice LC. Molecular phenotyping of human endometrium distinguishes menstrual cycle phases and underlying biological processes in normo-ovulatory women. Endocrinology. 2006;147:1097–121.CrossRef Talbi S, Hamilton AE, Vo KC, Tulac S, Overgaard MT, Dosiou C, Le Shay N, Nezhat CN, Kempson R, Lessey BA, Nayak NR, Giudice LC. Molecular phenotyping of human endometrium distinguishes menstrual cycle phases and underlying biological processes in normo-ovulatory women. Endocrinology. 2006;147:1097–121.CrossRef
59.
go back to reference Bulun SE, Cheng YH, Yin P, Imir G, Utsunomiya H, Attar E, Innes J, Julie Kim J. Progesterone resistance in endometriosis: link to failure to metabolize estradiol. Mol Cell Endocrinol. 2006;248:94–103.CrossRef Bulun SE, Cheng YH, Yin P, Imir G, Utsunomiya H, Attar E, Innes J, Julie Kim J. Progesterone resistance in endometriosis: link to failure to metabolize estradiol. Mol Cell Endocrinol. 2006;248:94–103.CrossRef
60.
go back to reference Ito K, Utsunomiya H, Yaegashi N, Sasano H. Biological roles of estrogen and progesterone in human endometrial carcinoma–new developments in potential endocrine therapy for endometrial cancer. Endocr J. 2007;54:667–79.CrossRef Ito K, Utsunomiya H, Yaegashi N, Sasano H. Biological roles of estrogen and progesterone in human endometrial carcinoma–new developments in potential endocrine therapy for endometrial cancer. Endocr J. 2007;54:667–79.CrossRef
61.
62.
go back to reference Feng YZ, Shiozawa T, Miyamoto T, Kashima H, Kurai M, Suzuki A, Ying-Song J, Konishi I. Overexpression of hedgehog signaling molecules and its involvement in the proliferation of endometrial carcinoma cells. Clin Cancer Res. 2007;13:1389–98.CrossRef Feng YZ, Shiozawa T, Miyamoto T, Kashima H, Kurai M, Suzuki A, Ying-Song J, Konishi I. Overexpression of hedgehog signaling molecules and its involvement in the proliferation of endometrial carcinoma cells. Clin Cancer Res. 2007;13:1389–98.CrossRef
63.
go back to reference Russell FA, King R, Smillie S-J, Kodji X, Brain SD. Calcitonin gene-related peptide: physiology and pathophysiology. Physiol Rev. 2014;94(4):1099–142.CrossRef Russell FA, King R, Smillie S-J, Kodji X, Brain SD. Calcitonin gene-related peptide: physiology and pathophysiology. Physiol Rev. 2014;94(4):1099–142.CrossRef
64.
go back to reference Zhao FP, Guo Z, Wang PF. Calcitonin gene related peptide (CGRP) inhibits norepinephrine induced apoptosis in cultured rat cardiomyocytes not via PKA or PKC pathways. Neurosci Lett. 2010;482:163–6.CrossRef Zhao FP, Guo Z, Wang PF. Calcitonin gene related peptide (CGRP) inhibits norepinephrine induced apoptosis in cultured rat cardiomyocytes not via PKA or PKC pathways. Neurosci Lett. 2010;482:163–6.CrossRef
65.
go back to reference Gomes RN, Castro-Faria-Neto HC, Bozza PT, Soares MB, Shoemaker CB, David JR, Bozza MT. Calcitonin gene-related peptide inhibits local acute inflammation and protects mice against lethal endotoxemia. Shock. 2005;24:590–4.CrossRef Gomes RN, Castro-Faria-Neto HC, Bozza PT, Soares MB, Shoemaker CB, David JR, Bozza MT. Calcitonin gene-related peptide inhibits local acute inflammation and protects mice against lethal endotoxemia. Shock. 2005;24:590–4.CrossRef
66.
go back to reference Wang X, Ebong SJ, Call DR, Newcomb DE, Bolgos GR, Remick DG. Calcitonin gene-related peptide partially reverses decreased production of chemokines KC and MIP-2 following murine sepsis. Inflammation. 2002;26:167–74.CrossRef Wang X, Ebong SJ, Call DR, Newcomb DE, Bolgos GR, Remick DG. Calcitonin gene-related peptide partially reverses decreased production of chemokines KC and MIP-2 following murine sepsis. Inflammation. 2002;26:167–74.CrossRef
67.
go back to reference Brain SD, Grant AD. Vascular actions of calcitonin gene-related peptide and adrenomedullin. Physiol Rev. 2004;84:903–34.CrossRef Brain SD, Grant AD. Vascular actions of calcitonin gene-related peptide and adrenomedullin. Physiol Rev. 2004;84:903–34.CrossRef
68.
go back to reference Brzozowski T, Konturek PC, Konturek SJ, et al. Ischemic preconditioning of remote organs attenuates gastric ischemia-reperfusion injury through involvement of prostaglandins and sensory nerves. Eur J Pharmacol. 2004;499:201–13.CrossRef Brzozowski T, Konturek PC, Konturek SJ, et al. Ischemic preconditioning of remote organs attenuates gastric ischemia-reperfusion injury through involvement of prostaglandins and sensory nerves. Eur J Pharmacol. 2004;499:201–13.CrossRef
Metadata
Title
Myometrial progesterone hyper-responsiveness associated with increased risk of human uterine fibroids
Authors
Mona Omar
Archana Laknaur
Ayman Al-Hendy
Qiwei Yang
Publication date
01-12-2019
Publisher
BioMed Central
Keyword
Progesterone
Published in
BMC Women's Health / Issue 1/2019
Electronic ISSN: 1472-6874
DOI
https://doi.org/10.1186/s12905-019-0795-1

Other articles of this Issue 1/2019

BMC Women's Health 1/2019 Go to the issue