THE BIOLOGY OF UTERINE FIBROIDS
Uterine fibroids (UFs), are the most common pelvic tumors, occurring in 70–80% of all reproductive-aged women and are the leading indication for hysterectomy worldwide.1–3 Although UFs are benign tumors, they typically cause severe menstrual bleeding, pelvic pain, preterm labor, recurrent abortion, and infertility. Hysterectomy is currently the main treatment used in women who no longer desire childbearing.4–6 UFs are hormonally responsive to estradiol and progesterone as well as other steroid hormones, and regress after menopause.7 Although, the cause of UFs is largely unknown, several risk factors are linked to UF development, which include age, race and ethnicity, family history, body mass index (BMI), etc.7,8
THE DEVELOPMENTAL ORIGINS OF HEALTH AND DISEASE
The Developmental Origins of Health and Disease (DOHaD) paradigm is one of the most rapidly growing areas of biomedical research now-a-days.9 This research field originated with early findings that prenatal nutrition was linked with late-onset coronary heart disease10 and malnutrition and low-level exposures to drugs and toxic substances are well tolerated by a pregnant woman, but her gestating fetus would be afflicted by adverse effects, some of which might become obvious only later in life.11,12 The field has now broadened to encompass a variety of environmental and occupational hazards. When these environmental insults disrupt early developmental processes, they may cause permanent changes in cellular characteristics that persist and then lead to increased susceptibility to a variety of diseases later in life.
Unlike in the adult, the perinatal/neonatal organ’s response to environmental exposure is much more rapid and severe.13,14 Environmental exposure is capable of causing organism toxicity due to immature immune system, lack of deoxyribonucleic acid (DNA) repair, poor liver metabolism, and incompletely formed organ barriers in early life stage. In addition to toxicity, environmental exposures during critical periods of organ development can permanently reprogram normal physiological responses to increase susceptibility to diseases later in life.15 The epigenomic programming that occurs during development exhibits a high degree of plasticity, and is modifiable by extrinsic factors such as environmental exposures.16,17
MOLECULAR MECHANISM OF REPROGRAMMING: ENVIRONMENTAL FACTORS ALTERS THE EPIGENOME OF UTERUS
Due to the plasticity during development of tissues and organs including uterine, accumulating evidence has shown that environmental factors act on epigenome via DNA methylation and histone modification, which eventually lead to alteration of gene expression pattern and related diseases later in life. In utero the exposure of bisphenol A (BPA), an organic synthetic compound belonging to the group of diphenylmethane derivatives and bisphenols, altered the global CpG methylation profile of the uterine genome and subsequent gene expression pattern. Changes in estrogen response were accompanied by altered methylation that preferentially affected estrogen receptor-α (ERα)-binding genes.18 Neonatal exposure of CD-1 mice to diethylstilbestrol (DES), a synthetic non-steroidal estrogen of the stilbestrol group, induced uterine adenocarcinoma in aging animals, concomitantly decreasing DNA methylation of nucleosome binding protein 1 (Nsbp1) promoter CpG Island (CGI) in the uteri which leads to persistent overexpression of Nsbp1 throughout life.16,19 Moreover, 17β-estradiol and other environmental estrogens (DES and genistein) are capable of inducing phosphoinositide 3-kinase (PI3K)/AKT non-genomic estrogen receptor signaling to the histone EZH2,20 and therefore reduced levels of trimethylation of lysine 27 on histone H3 in hormone-responsive cells.20,21 These studies provide a direct link between xenoestrogen-induced nuclear hormone receptor signaling and modulating of epigenetic machinery in response to environmental estrogen in UFs.
EPIGENETIC REPROGRAMMING OF STEM CELLS IN UTERINE FIBROIDS IN RESPONSE TO EARLY-LIFE EXPOSURES TO ENDOCRINE DISRUPTING CHEMICALS
For environmental diseases, a central subject to resolve is the role of stem cells in the tumorigenesis or pre-cursors of degenerative diseases. Developmental adverse exposures may affect the highly regulated differentiation of hematopoietic stem cells, and even slight changes in the feature of these cells may serve as indications of health effects that may not be observed until later in life and may be magnified during the entire life.22 The environmental exposure directs the behavior of stem and progenitor cells, the fundamental source from which all tissues derive. However, environmental health studies are lacking on stem cells.17 UF growth and progression depend on a specialized subpopulation of tumor cells, termed tumor initiating cells (TICs).3,23 Thus, TICs represent a critical therapeutic target, but the molecular mechanisms that regulate them are poorly understood.
To determine the mechanism underlying increased risk of UF development at stem cell levels, we have recently determined the effect of early-life exposure to endocrine disrupting chemicals (EDCs) on stem cell behavior as well as characterized myometrial stem cells (MSCs) as a target for ethnic and environmental factors that increase UF risk. We utilized Eker rats carrying a germ-line mutation in the tuberous sclerosis complex 2 (Tsc2) tumor suppressor gene, that are susceptible for development of UFs which share similar anatomic, histologic, and biologic features to human UFs.24 Using this model, we isolated and characterized Stro1+/CD44+ MSC/progenitor-like cells that give rise to UFs, which resided in the rat cervix, a hypoxic niche in the uterus.25 These Stro-1+/CD44+ MSCs responded to environmental cues, and expanded in response to developmental environmental exposures that promote UF development.25
Human female reproductive tract has been shown to be a target for developmental programming as a result of inappropriate early life hormone exposure.17,26 Early life exposure to EDC compounds have been connected to increased risk of adult onset of UFs in women.27,28 Minority communities are particularly at risk for hazardous environmental exposures.29–31 However, similar inquiries in humans are lacking due to difficulties in collecting suitable human myometrial samples and/or to ascertain environmental exposures. We proposed that myometrium from a non-fibroid uterus that does not exhibit any detectable myometrial pathology (removed for benign gynecological indications such as pelvic organ prolapsed near the end of reproductive life≥45 years) would model normal myometrium (MyoN). In turn, myometrium collected from fibroid uteri, would model for at risk myometrium or myometrium that might be exposed in early life to injurious environmental hormonal disruptors or other toxins (MyoF). We collect these MyoF tissues at least 2 cm away from the closest fibroid to avoid any potential mechanical or paracrine effect of the fibroid tumor itself. We demonstrated by immunohistochemistry analysis that MyoF samples from African American and Caucasian, consistently exhibited significantly higher numbers of Stro1+/CD44+ MSCs as compared to age-, BMI-, and menstrual cycle phase-matched MyoN tissues.25
Although the role of MSCs in development of UFs is extremely important,23,32,33 the molecular mechanism underlying developmental exposure to EDCs and other toxins at MSCs levels has not been characterized before.15 By ribonucleic acid (RNA)- sequencing analysis, we recently identified some key genes including estrogen responsive genes (ERGs) that are differentially regulated in MSCs early-life exposed to diethylstilbestrol (DES) verse control (VEH).34 Subsequently, we performed gene set enrichment analysis on the ChIP-sequencing data and found enrichment of histone H3 trimethylated at lysine 4 (H3K4me3) (an active mark for gene transcription) at the promoters of ERGs in DES-MSCs as compared to VEH-MSCs. Furthermore, the increased expression of ERGs in DES-MSCs was positively correlated with the elevated H3K4me3 epigenetic mark.34 Our current study suggest that early life exposure to DES during sensitive periods of uterine development increases the risk of UF development by reprogramming the epigenome of MSCs towards a pro-fibroid epigenomic landscape. Further understandings of EDC-induced epigenetic alteration and DNA mutations in MSCs have the potential to substantially advance UF research.
Acknowledgments
This work was supported in part by an Augusta University Startup package, the National Institutes of Health grant HD04622811 (to AA), and the Augusta University Intramural Grants Program (QY).
Footnotes
CONFLICTS OF INTEREST
None of the authors have a financial relationship with a commercial entity that has an interest in the subject of this manuscript.
References
- 1.Al-Hendy A, Salama S. Gene therapy and uterine leiomyoma: A review. Hum Reprod Update. 2006;12(4):385–400. doi: 10.1093/humupd/dml015. [DOI] [PubMed] [Google Scholar]
- 2.Bulun SE. Uterine fibroids. N Engl J Med. 2013;369:1344–1355. doi: 10.1056/NEJMra1209993. [DOI] [PubMed] [Google Scholar]
- 3.Yang Q, Mas A, Diamond MP, Al-Hendy A. The mechanism and function of epigenetics in uterine leiomyoma development. Reprod Sci. 2016;23(2):163–175. doi: 10.1177/1933719115584449. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Segars JH, Parrott EC, Nagel JD, et al. Proceedings from the Third National Institutes of Health International Congress on Advances in Uterine Leiomyoma Research: Comprehensive review, conference summary and future recommendations. Hum Reprod Update. 2014;20:309–333. doi: 10.1093/humupd/dmt058. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Sheiner E, Bashiri A, Levy A, Hershkovitz R, Katz M, Mazor M. Obstetric characteristics and perinatal outcome of pregnancies with uterine leiomyomas. J Reprod Med. 2004;49(3):182–186. [PubMed] [Google Scholar]
- 6.Hart R, Khalaf Y, Yeong CT, Seed P, Taylor A, Braude P. A prospective controlled study of the effect of intramural uterine fibroids on the outcome of assisted conception. Hum Reprod. 2001;16(11):2411–2417. doi: 10.1093/humrep/16.11.2411. [DOI] [PubMed] [Google Scholar]
- 7.Flake GP, Andersen J, Dixon D. Etiology and pathogenesis of uterine leiomyomas: A review. Environ Health Perspect. 2003;111(8):1037–1054. doi: 10.1289/ehp.5787. Web site. https://www.ncbi.nlm.nih.gov/pmc/articles/PMC1241553/. Accessed November 28, 2016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Laughlin SK, Schroeder JC, Baird DD. New directions in the epidemiology of uterine fibroids. Semin Reprod Med. 2010;28(3):204–217. doi: 10.1055/s-0030-1251477. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Grandjean P, Barouki R, Bellinger DC, et al. Life-Long implications of developmental exposure to environmental stressors: New perspectives. Endocrinology. 2015;156:3408–3415. doi: 10.1210/EN.2015-1350. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Barker DJ, Osmond C. Infant mortality, childhood nutrition, and ischaemic heart disease in England and Wales. Lancet. 1986;1:1077–1081. doi: 10.1016/S0140-6736(86)91340-1. [DOI] [PubMed] [Google Scholar]
- 11.Barouki R, Gluckman PD, Grandjean P, Hanson M, Heindel JJ. Developmental origins of non-communicable disease: Implications for research and public health. Environmental Health: A Global Access Science Source. 2012;11:42. doi: 10.1186/1476-069X-11-42. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Heindel JJ, Balbus J, Birnbaum L, et al. Developmental origins of health and disease: Integrating environmental influences. Endocrinology. 2015;156(10):3416–3421. doi: 10.1210/EN.2015-1394. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Yang Q, Sun M, Ramchandran R, Raj JU. IGF-1 signaling in neonatal hypoxia-induced pulmonary hypertension: Role of epigenetic regulation. Vascul Pharmacol. 2015;73:20–31. doi: 10.1016/j.vph.2015.04.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Sun M, Ramchandran R, Chen J, Yang Q, Raj JU. Smooth muscle insulin-like growth factor-1 mediates hypoxia-induced pulmonary hypertension in neonatal mice. Am J Respir Cell Mol Biol. 2016 doi: 10.1165/rcmb.2015-0388OC. [In Press] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Yang Q, Diamond MP, Al-Hendy A. Early life adverse environmental exposures increase the risk of uterine fibroid development: Role of epigenetic regulation. Front Pharmacol. 2016;7:40. doi: 10.3389/fphar.2016.00040. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Walker CL, Ho SM. Developmental reprogramming of cancer susceptibility. Nat Rev Cancer. 2012;12(578):479–486. doi: 10.1038/nrc3334. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Katz TA, Yang Q, Trevino LS, Walker CL, Al-Hendy A. Endocrine-disrupting chemicals and uterine fibroids. Fertil Steril. 2016;106:967–977. doi: 10.1016/j.fertnstert.2016.08.023. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Jorgensen EM, Alderman MH, 3rd, Taylor HS. Preferential epigenetic programming of estrogen response after in utero xenoestrogen (bisphenol-A) exposure. FASEB J. 2016;30(9):3194–3201. doi: 10.1096/fj.201500089R. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Gibson DA, Saunders PT. Endocrine disruption of oestrogen action and female reproductive tract cancers. Endocr Relat Cancer. 2014;21:T13–T31. doi: 10.1530/ERC-13-0342. [DOI] [PubMed] [Google Scholar]
- 20.Bredfeldt TG, Greathouse KL, Safe SH, Hung MC, Bedford MT, Walker CL. Xenoestrogen-induced regulation of EZH2 and histone methylation via estrogen receptor signaling to PI3K/AKT. Mol Endocrinol. 2010;24(5):993–1006. doi: 10.1210/me.2009-0438. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Greathouse KL, Bredfeldt T, Everitt JI, et al. Environmental estrogens differentially engage the histone methyltransferase EZH2 to increase risk of uterine tumorigenesis. Mol Cancer Res. 2012;10(4):546–557. doi: 10.1158/1541-7786.MCR-11-0605. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Laiosa MD, Tate ER. Fetal hematopoietic stem cells are the canaries in the coal mine that portend later life immune deficiency. Endocrinology. 2015;156:3458–3465. doi: 10.1210/en.2015-1347. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Ono M, Bulun SE, Maruyama T. Tissue-specific stem cells in the myometrium and tumor-initiating cells in leiomyoma. Biol Reprod. 2014;91(6):149. doi: 10.1095/biolreprod.114.123794. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Walker CL, Hunter D, Everitt JI. Uterine leiomyoma in the Eker rat: A unique model for important diseases of women. Genes, Chromosomes Cancer. 2003;38(4):349–356. doi: 10.1002/gcc.10281. [DOI] [PubMed] [Google Scholar]
- 25.Mas A, Stone L, O’Connor PM, et al. Developmental exposure to endocrine disruptors expands murine myometrial stem cell compartment as a prerequisite to leiomyoma tumorigenesis. Stem Cells. 2016 doi: 10.1002/stem.2519. [In Press] [DOI] [PubMed] [Google Scholar]
- 26.Prusinski L, Al-Hendy A, Yang Q. Developmental exposure to endocrine disrupting chemicals alters the epigenome: Identification of reprogrammed targets. Gynecol Obstet Res. 2016;3(1):1–6. doi: 10.17140/GOROJ-3-127. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Wise LA, Palmer JR, Reich D, Cozier YC, Rosenberg L. Hair relaxer use and risk of uterine leiomyomata in African-American women. Am J Epidemiol. 2012;175:432–440. doi: 10.1093/aje/kwr351. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.D’Aloisio AA, Baird DD, DeRoo LA, Sandler DP. Association of intrauterine and early-life exposures with diagnosis of uterine leiomyomata by 35 years of age in the Sister Study. Environ Health Perspect. 2010;118:375–381. doi: 10.1289/ehp.0901423. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Silbergeld EK, Patrick TE. Environmental exposures, toxicologic mechanisms, and adverse pregnancy outcomes. Am J Obstet Gynecol. 2005;192:S11–S21. doi: 10.1016/j.ajog.2004.06.117. [DOI] [PubMed] [Google Scholar]
- 30.Weintraub M, Birnbaum LS. Catfish consumption as a contributor to elevated PCB levels in a non-Hispanic black subpopulation. Environ Res. 2008;107(3):412–417. doi: 10.1016/j.envres.2008.03.001. [DOI] [PubMed] [Google Scholar]
- 31.Meadows M. Heading off hair-care disasters. Use caution with relaxers and dyes. FDA Consumer. 2001;35:21–24. Web site. http://permanent.access.gpo.gov/lps1609/www.fda.gov/fdac/features/2001/101_hair.html. Accessed November 28, 2016. [PubMed] [Google Scholar]
- 32.Bulun SE, Moravek MB, Yin P, et al. Uterine leiomyoma stem cells: Linking progesterone to growth. Semin Reprod Med. 2015;33(5):357–365. doi: 10.1055/s-0035-1558451. [DOI] [PubMed] [Google Scholar]
- 33.Mas A, Nair S, Laknaur A, Simon C, Diamond MP, Al-Hendy A. Stro-1/CD44 as putative human myometrial and fibroid stem cell markers. Fertil Steril. 2015;104(1):225–234.e3. doi: 10.1016/j.fertnstert.2015.04.021. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Yang Q, Trevino L, Mas A, et al. Early life developmental exposure to endocrine disrupting chemicals increases the risk of adult onset of uterine fibroids by permanently reprograming the epigenome of myometrial stem cells towards a pro-fibroid landscape. Fertil Steril. 2016;106(3):e2. doi: 10.1016/j.fertnstert.2016.07.012. [DOI] [Google Scholar]