Skip to main content

5-aza-2′-deoxycitidine inhibits cell proliferation, extracellular matrix formation and Wnt/β-catenin pathway in human uterine leiomyomas

Abstract

Background

Uterine leiomyoma is a benign tumor with unclear pathogenesis and inaccurate treatment. This tumor exhibits altered DNA methylation related to disease progression. DNMT inhibitors as 5-aza-2′-deoxycytidine (5-aza-CdR), have been suggested to treat tumors in which DNA methylation is altered. We aimed to evaluate whether DNA methylation reversion with 5-aza-CdR reduces cell proliferation and extracellular matrix (ECM) formation in uterine leiomyoma cells to provide a potential treatment option.

Methods

Prospective study using uterine leiomyoma and adjacent myometrium tissues and human uterine leiomyoma primary (HULP) cells (n = 16). In tissues, gene expression was analyzed by qRT-PCR and DNMT activity by ELISA. Effects of 5-aza-CdR treatment on HULP cells were assessed by CellTiter, western blot, and qRT-PCR.

Results

DNMT1 gene expression was higher in uterine leiomyoma vs myometrium. Similarly, DNMT activity was greater in uterine leiomyoma and HULP cells (6.5 vs 3.8 OD/h/mg; 211.3 vs 63.7 OD/h/mg, respectively). After 5-aza-CdR treatment on HULP cells, cell viability was reduced, significantly so at 10 μM (85.3%). Treatment with 10 μM 5-aza-CdR on HULP cells significantly decreased expression of proliferation marker PCNA (FC = 0.695) and of ECM proteins (COLLAGEN I FC = 0.654; PAI-1, FC = 0.654; FIBRONECTIN FC = 0.733). 5-aza-CdR treatment also decreased expression of Wnt/β-catenin pathway final targets, including WISP1 protein expression (10 μM, FC = 0.699), c-MYC gene expression (2 μM, FC = 0.745 and 10 μM, FC = 0.728), and MMP7 gene expression (5 μM, FC = 0.520 and 10 μM, FC = 0.577).

Conclusions

5-aza-CdR treatment inhibits cell proliferation, ECM formation, and Wnt/β-catenin signaling pathway targets in HULP cells, suggesting that DNA methylation inhibition is a viable therapeutic target in uterine leiomyoma.

Background

Uterine leiomyomas (UL) are monoclonal benign tumors originating from smooth muscle cells located in the myometrium (MM) that affect to 25–30% of women of reproductive age [1, 2]. Prevalence of uterine leiomyomas in African American women is three times higher than Caucasian women [3, 4]. Around 30% of patients with leiomyomas present symptoms such as excessive uterine bleeding, anemia, pelvic pain, infertility, recurrent pregnancy loss, and/or preterm birth [5]. Although the gold-standard treatment for leiomyoma is surgical myomectomy or hysterectomy, other less invasive hormonal treatments, such as gonadotropin releasing-hormone agonist (aGnRH) [6] or ulipristal acetate (UPA) have been used to treat leiomyomas [7]. However, these treatments present side effects such as menopausal symptoms or hepatic damage [8], and once the treatment is stopped, leiomyomas enlarge again, in most cases recovering their initial size after 6 months [9]. For this reason, there is not an effective therapy to treat uterine leiomyoma with minimal side effects, despite being the most prevalent gynecological tumor.

Although pathogenesis of leiomyomas remains unclear, many factors have been proposed to contribute in the development of uterine leiomyomas. These factors include steroid hormones such as estrogens [10] and progesterone [11]; growth factors such as tumor growth factor beta (TGF-β), vascular endothelial growth factor (VEGF), acidic fibroblast growth factor (FGF), and basic FGF (bFGF) [12]; alterations of the Wnt/β-catenin pathway [13]; and genetic and epigenetic alterations [14]. Different subtypes of genetic mutations are described as a possible cause of leiomyoma development, including high mobility group AT-hook 2 (HMGA2) rearrangements, mediator complex subunit 12 (MED12) mutations, biallelic inactivation of fumarate hydratase (FH), and deletions affecting collagen type IV alpha 5 and alpha 6 (COL4A5 and COL4A6) [15]. In addition, some environmental factors increase risk for developing uterine leiomyomas; these include high body mass index (BMI), meat consumption, alcohol consumption, hypertension, and vaginal infections [16,17,18]. These data suggest that epigenetic factors are also involved in the development of uterine leiomyoma. While genetic mutations are difficult to reverse, some epigenetic changes can be reversed, for example by chemical agents. This possibility suggests that there could be new therapeutic options for patients with UL.

DNA methylation is a widely studied epigenetic mark that consists of the addition of a methyl group to the 59-carbon of the cytosine ring through a covalent union within the CpG island after replication [19]. Specifically, methylation of a promoter CpG island triggers binding of proteins that condense the chromatin and make the promoter region inaccessible to transcription factors, blocking transcription initiation and, thereby, repressing gene expression [20]. DNA methylation is catalyzed by DNA methyltransferases (DNMT), of which there are three main types: DNMT1, DNMT3A, and DNMT3B [14]. DNMT1 maintains DNA methylation patterns during DNA replication; in contrast, DNMT3A and DNMT3B establish new methylation patterns and are therefore called de novo methyltransferases [21]. Aberrant DNA methylation, through oncogene hypo-methylation and tumor suppressor gene hyper-methylation, is involved in tumorigenesis of various cancer types, such as prostate cancer or neuroblastoma [22,23,24,25]. In UL, some studies reported differential expression of DNMTs and an aberrant DNA methylation compared with the adjacent MM, along with repression of tumor suppressor genes and other genes involved in cell cycle regulation, cell growth, migration, and extracellular matrix (ECM) formation [26,27,28,29,30]. Of particular interest is the Wnt/β-catenin pathway, which regulates proliferation, survival, migration and differentiation of many cell types and is dysregulated in numerous cancers [31,32,33,34] and uterine leiomyomas cells [7, 13, 35,36,37]. There is potential that aberrant DNA methylation affects this pathway in UL as it does in other tumor types, such as colorectal cancer [38]. For other tumors with altered DNA methylation that is related to disease progression, the use of DNMT inhibitors such as 5-aza-2′-deoxycytidine (5-aza-CdR) can offer effective treatment. 5-aza-CdR is a nucleoside analog that acts as a DNA demethylating agent through inhibition of DNMTs (DNMT1/3A/3B) [19], leading to changes in gene reactivation. 5-aza-CdR has been demonstrated that induce cell cycle arrest, inhibition of cell differentiation, and cell death by inhibiting post-replication methylation of DNA in human and mouse endometrium [39, 40]. Given the altered DNA methylation patterns and DNMT expression in UL, demethylation of human uterine leiomyoma cells by this DNMT inhibitor could reduce leiomyoma size, offering a new therapeutic option. However, the effect of 5-aza-CdR on UL cells is not well studied; further research is necessary to determine how this inhibitor affects the regulation of molecular mechanisms involved in leiomyoma development.

Here, we evaluated the effect of 5-aza-CdR in human uterine leiomyoma primary (HULP) cells on cell proliferation, apoptosis, ECM formation, and the Wnt/β-catenin pathway to test its potential as a new therapeutic option for leiomyoma.

Methods

Human tissue collection

Human UL and adjacent MM were collected from premenopausal women aged 31–48 years undergoing myomectomy or hysterectomy due to symptomatic UL pathologies (n = 16) without any previous hormonal treatment for the last 3 months (Supplemental Table 1).

Ethical approval

This study was approved by Clinical Ethics Committee at Hospital Univeristario y Politecnico La Fe (Spain) (2018/0097), and all participants provided informed consent.

Human uterine leiomyoma primary cell isolation

UL and MM fragments were mechanically dissected into small pieces that were incubated at 37 °C with 2 mg/mL type II collagenase (Labclinics, Spain) and 1 mg/mL DNase I (Sigma-Aldrich, Saint Louis, MO) to obtain single-cell suspensions. Subsequently, cells were filtered through 50-μm polyethylene filters (Partec, Celltrics) to remove cellular clumps and undigested tissue. Isolated cells were incubated in vitro at 37 °C and 5% CO2 in culture medium [Dulbecco’s Modified Eagle Medium (DMEM)/F-12 (Gibco, Waltham, MA) with 10% fetal bovine serum (FBS) and antibiotic-antimycotic solution] for subsequent experiments of DNMT activity and 5-aza-CdR treatment.

Study of DNMT activity in vivo and in vitro

To evaluate DNMT activity over time in HULP and MM cells isolated from UL and adjacent MM tissue (n = 3), cells were incubated in culture medium for 7, 9, or 10 days. Subsequently, HULP and MM cells were collected, and nuclear protein extracts were isolated using EpiQuik Nuclear Extraction Kit (Epigentek, Brooklyn, NY), which was also used to obtain nuclear proteins from UL and MM tissues (n = 7). DNMT activity in tissues and in vitro cultured HULP and MM cells was measured with EpiQuik DNMT Activity/Inhibition ELISA Easy Kit immunoassay (Epigentek, Brooklyn, NY) using a microplate reader (Synergy HT, Bio-Tek). The ratio of methylated DNA was measured at a wavelength of 450 nm. The activity of DNMT enzymes, which is proportional to the optical density (OD) intensity measured, was calculated according to the formula provided by the manufacturer:

$$ DNMT\kern0.34em Activity\kern0.28em \left( OD/h/ mg\right)=\frac{\left( Sample\kern0.28em OD- Blank\kern0.28em OD\right)}{\left( Protein\kern0.34em amount\kern0.28em \left(\mu g\right)\times hour\right)}\times 100 $$

5-aza-2′-deoxycitidine treatment

To determine the effect of 5-aza-CdR on HULP cells, cells were incubated in culture medium (day 0 = D0) until achieving a confluence of 70% (day 3 = D3). Then, they were starved in serum-free medium overnight and treated (day 4 = D4) with different doses of 5-aza-dC (Abcam, Cambridge, UK) (n = 8): 0 μM (control), 2 μM (low-dose), 5 μM 5-aza-CdR (middle-dose), 10 μM (high-dose) for 72 h (day 7 = D7). Medium was changed every 24 h.

Cell viability assay

HUPL cells (n = 16) were cultured in 96-well plate in culture medium; once 70% of confluence was achieved, cells were starved in serum-free medium overnight and treated (D4) with 0, 2, 5, or 10 μM of the DNMT inhibitor 5-aza-CdR for 72 h (D7). Medium was changed every 24 h. After this period, the quantity of viable cells in proliferation was measured with CellTiter 96 AQueous One Solution Cell Proliferation Assay (Promega, Madison, WI) and absorbance was measured on a microplate reader (Synergy HT, Bio-Tek) at 490 nm.

Protein extraction and western blot analysis

Proteins from HULP cells (n = 8) treated with/without 5-aza-CdR were extracted using radioimmunoprecipitation assay (RIPA) buffer containing protease inhibitors. Subsequently, HULP cells lysates (10 μg to 20 μg of protein) were analyzed by 12% sodium dodecyl sulphate-polyacrylamide gel electrophoresis (SDS-PAGE) and then transferred to PVDF membranes. Western blot (WB) analysis was conducted to measure expression of ECM protein Plasminogen activator inhibitor-1 (PAI-I; sc-5297) (1:200), proliferating cell nuclear antigen (PCNA; sc-56) (1:200), Wnt1-inducible-signaling pathway protein 1 (WISP1; sc-133,126) (1:200), B-cell lymphoma-2 (BCL2; sc-7382) (1:200), and BCL2 associated-X (BAX; sc-20,067) (1:200) from Santa Cruz Biotechnology (Santa Cruz, CA). Expression of extracellular matrix proteins COLLAGEN I (COL-I;70R-CR007X) (1:1000, Fitzgerald, Acton, MA) and FIBRONECTIN (F3648) (1:2000, Sigma-Aldrich) was also evaluated. Antigen-antibody complex was detected with SuperSignal West Femto Maximun Sensitivity Substrate (ThermoFisher, Waltham, MA) and specific protein bands were visualized by chemiluminescence imaging using the LAS-3000 Imaging-System (Fujifilm, Tokyo, Japan). The intensity of each protein band was quantified with ImageJ software (National Institutes of Health, Bethesda, MD) and normalized in relation to its corresponding housekeeping protein, β-actin (1:1000; sc-47,778).

Gene expression analysis: DNMT and Wnt/β-catenin pathway

Total RNA from UL and MM tissues was extracted with TRIzol reagent (Fisher Scientific, Waltham, MA) for the study of DNMT1 expression. Total RNA from HULP cells treated with/without 5-aza-CdR was obtained using RNeasy Mini kit (Qiagen, Hilden, Germany) to assess expression of final targets in the Wnt/B-catenin pathway. cDNA was synthesized using PrimeScript RT reagent Kit (Takara, Japan). Quantitative real-time polymerase chain reaction (qRT-PCR) was performed with StepOnePlus System (Applied Biosystems, Foster City, CA) using PowerUp SYBR Green (Fisher Scientific, Waltham, MA). Gene expression of final targets of Wnt pathway, MYC proto-oncogene (c-MYC) and Matrix Metallopeptidase 7 (MMP7), was analyzed in HULP cells treated with and without 5-aza-CdR (n = 8). In addition, DNMT1 expression was analyzed in UL and MM tissues (n = 8). Data were normalized with Glyceraldehyde-3-Phosphate Dehydrogenase (GAPDH), a housekeeping gene with minor variability. Primers were designed using Primer Quest Tool (DNA Integrated Technologies, Coralville,IA) (Supplemental Table II). ΔΔCt method was used to calculate fold change.

Statistical analysis

GraphPad Prism 8.0 was used for statistical analyses and graphic generation (San Diego, CA). Normality and logarithmic tests (Anderson-Darling, D’Agostino & Pearson, Shapiro-Wilk and Kolmogorov-Smirnov test) were performed to analyze the distribution of our results. A paired t-test was performed for DNMT activity study in UL vs MM tissue as well as HULP and MM cells. The Wilcoxon test for paired samples was performed to analyze qRT-PCR results of DNMT1 gene expression in UL tissue and adjacent MM. Repeated measures one-way ANOVA test with the Geisser-Greenhouse correction was performed for qRT-PCR analysis of multicomponent variants of c-MYC and MMP7; for WB analysis of WISP1, PCNA, and COL-I; and for cell viability assay of HULP and MM cells. Friedmann test was used to analyze BAX-BCL2 ratio, PAI-I, and FIBRONECTIN. Results of 5-aza-CdR treatment were normalized and compared with control HULP cells without treatment. Data are presented as mean ± standard deviation (SD). p-value < 0.05 was considered statistically significant.

Results

DNTM1 gene expression and DNMT activity in UL vs MM tissues

To examine whether increased global DNA methylation previously observed in UL vs MM was due to a higher expression of DNMT enzymes, DNMT1 gene expression was analyzed by qRT-PCR in UL and MM tissues. UL tissue showed a statistically significant higher gene expression of DNMT1 compared to MM tissue (fold change = 2.49, p-value = 0.0295) (Fig. 1A). Furthermore, DNMT activity was measured by ELISA in UL vs MM tissues to determine the correlation between gene expression and DNMT activity. Significantly higher DNMT activity was observed in UL tissue compared to adjacent MM tissue (6.50 vs 3.76 OD/h/mg, p-value = 0.026) (Fig. 1B).

Fig. 1
figure 1

DNMT status in UL compared with adjacent MM. A Gene expression levels of DNMT1 in UL compared to MM tissues (n = 14) represented as fold change. B DNMT activity (OD/h/mg) of UL compared to MM tissues (n = 7). C Effects of cell culture on DNA methyltransferase (DNMT) activity of HULP and MM cells (n = 3) cultured for 7 days, 9 days, and 10 days. Data are expressed as the mean value ± standard error (SEM). * p-value < 0.05

DNMT activity in HULP and myometrial cells in vitro

Since epigenetic changes depend on the environment, experiments were conducted to determine the correlation between DNMT activity in vivo in UL and adjacent MM tissue and HULP and MM cells cultured in vitro. For this purpose, DNMT activity of HULP and MM cells was measured at 7, 9, and 10 days of in vitro culture. DNMT activity was greater in HULP cell compared to MM cells at day 7 (211.30 vs 63.67 OD/h/mg, p-value = 0.284), day 9 (66.41 vs 11.88 OD/h/mg, p-value = 0.217), and day 10 (24.75 vs 6.47 OD/h/mg, p-value = 0.337) (Fig. 1C). These results corroborated that the increased DNMT activity observed in UL compared to MM tissues is not modified in vitro by cell culture conditions (Fig. 1C).

In vitro effects of 5-aza-2′-deoxycytidine on cell proliferation in HULP cells

To demonstrate the antiproliferative effect of 5-aza-CdR on HULP cells, we measured the number of proliferating cells. Treatment with 5-aza-CdR decreased the percentage of viable HULP cells in a dose-dependent manner, being this decrease statistically significant with high-dose (10 μM) treatment in HULP cells (93.93% at 2 μM, p-value = 0.114; 92.15% at 5 μM, p-value = 0.053; 85.25% at 10 μM, p-value = 0.0001) compared to 0 μM (Fig. 2A).

Fig. 2
figure 2

Effect of 5-aza-CdR treatment on cell survival in HULP cells. A Percentage of viable HULP (n = 16) cells after 72 h of treatment with 0, 2, 5, or 10 μM of 5-aza-CdR. B Representative images of protein expression levels for BCL2 (23 kDa) and BAX (26 kDa) and C PCNA (36 kDa) analyzed in HULP (n = 8) after treatment with 5-aza-CdR at 0, 2, 5, or 10 μM (D-E) Means and standard deviations of normalized data of BAX/BCL2 ratio and PCNA, both represented as fold-change. * p-value < 0.05 *** p-value < 0.001

Subsequently, to clarify whether this decrease in viable cells was due to cell death increase and/or to cell proliferation decrease, protein markers of apoptosis status and proliferation were analyzed in 5-aza-CdR-treated HULP cells by western blot. We measured BAX (proapoptotic) and BCL2 (anti-apoptotic) protein expression (Fig. 2B) and did not observe an increased apoptosis (no BCL2 upregulation or BAX downregulation). Moreover, we calculated the BAX/BCL2 ratio to determine the susceptibility to apoptosis in 5-aza-CdR-treated HULP cells compared with HULP cells without treatment (0 μM). Although the BAX/BCL2 ratio showed a trend to increase in HULP cells after treatment, no significant differences were observed with 2, 5, or 10 μM of 5-aza-CdR [fold change (FC) = 1.46, p-value = 0.099; FC = 1.54, p-value = 0.158; FC = 1.13, p-value = 0.099, respectively) compared to untreated cells (Fig. 2D). Therefore, 5-aza-CdR-treated HULP cells did not show a significant change in apoptosis. To further analyze the decrease in cell viability observed in 5-aza-CdR-treated HULP cells, we assessed effect of 5-aza-CdR on cell proliferation by analyzing protein expression of the gold-standard proliferation marker PCNA using western blot in 5-aza-CdR-treated and untreated HULP cells (Fig. 2C). 5-aza-CdR treatment decreased PCNA protein expression in HULP cells in a dose-dependent manner, reaching statistical significance at the high dose (2 μM FC = 0.877, p-value = 0.099; 5 μM FC = 0.794, p-value = 0.055; 10 μM FC = 0.695, p-value = 0.034, respectively) (Fig. 2E).

In vitro effects of 5-aza-2′-deoxycytidine on extracellular matrix in HULP cells

To evaluate ECM status in HULP cells after 5-aza-CdR treatment, expression of proteins involved in ECM formation, such as COLLAGEN I, FIBRONECTIN, and PAI-1, was determined by western blot (Fig. 3A, B, and C, respectively). 5-aza-CdR treatment decreased COLLAGEN I expression in a dose-dependent manner, reaching statistical significance at 10 μM (FC = 0.654, p-value = 0.023; Fig. 3D). In addition, a tend to decrease COLLAGEN I was observed at 2 μM and 5 μM (FC = 0.6540.897, p-value = 0.729; FC = 0.675, p-value = 0.104, respectively) (Fig. 3D). Although no changes were found in FIBRONECTIN expression at 5 μM (FC = 0.826, p-value = 0.244), a statistically significant reduction was observed at 2 μM (FC = 0.812, p-value = 0.020) and 10 μM (FC = 0.733, p-value = 0.035) (Fig. 3E). Finally, PAI-1 expression was significantly decreased compared to control group (0 μM) after 5-aza-CdR treatment at 5 μM and 10 μM (FC = 0.865, p-value = 0.035; FC = 0.766, p-value = 0.020, respectively) (Fig. 3F). No statistically significant differences were found at 2 μM 5-aza-CdR (FC = 0.885, p-value = 0.244). These results showed that 5-aza-CdR inhibited ECM protein expression in HULP cells in vitro in a dose-dependent manner.

Fig. 3
figure 3

Extracellular matrix evaluation in HULP cells after 5-aza-CdR treatment. Representative images of protein expression levels for A COLLAGEN I (140 kDa), B FIBRONECTIN (220 kDa), and C PAI-1 (50 kDa) and normalized quantitative protein expression of D COLLAGEN I, E FIBRONECTIN, and F PAI-1 in the HULP cells at different treatment groups with 5-aza-CdR at 0, 2, 5, or 10 μM for 72 h (n = 8). Data are represented as mean and deviations of fold change. * p-value < 0.05

In vitro effects of 5-aza-2′-deoxycytidine on Wnt/β-catenin signaling pathway in HULP cells

To assess the effect of 5-aza-CdR treatment on Wnt/β-catenin pathway on HULP cells, we measured the protein expression of WISP1, a Wnt/β-catenin target protein, in HULP cells in presence and absence of 5-aza-CdR treatment by western blot. Results revealed an inhibition of WISP1 protein expression in HULP cells treated with 5-aza-CdR compared to control group (0 μM) in a dose-dependent manner treatment (Fig. 4A), reaching statistical significance at 10 μM (2 μM FC = 0.903, p-value = 0.408; 5 μM FC = 0.860, p-value = 0.071; 10 μM FC = 0.699, p-value = 0.026, respectively) (Fig. 4B).

Fig. 4
figure 4

Wnt/β-catenin signaling pathway analysis in HULP cells after DNMT inhibition with 5-aza-CdR treatment. A Representative images of protein expression for WISP1 in HULP cells after treatment with 5-aza-CdR at 0, 2, 5, or 10 μM for 72 h. B Normalized quantitative protein expression of WISP1 represented as mean and deviations of fold-change. Gene expression levels of C c-MYC and D MMP7 in HULP cells treated for 72 h with 2, 5, or 10 μM of 5-aza-CdR compared to untreated HULP cells (0 μM), represented as fold-change. * p-value < 0.05 ** p-value < 0.01

To further analyze the regulation of Wnt/β-catenin signaling pathway by 5-aza-CdR treatment in HULP cells, gene expression levels of c-MYC and MMP7, as final targets of Wnt/β-catenin pathway, were determined in these cells by qRT-PCR. c-MYC was significantly downregulated in HULP cells after 5-aza-CdR treatment at all doses tested compared to HULP cells without treatment, reaching significance at 2 μM and 10 μM doses (2 μM FC = 0.745, p-value = 0.028; 10 μM FC = 0.728, p-value = 0.019, respectively) (Fig. 4C), while no significant differences were found at 5 μM (FC = 0.810, p-value = 0.287). Finally, a significantly decrease of MMP7 expression was observed in HULP cells after 5-aza-CdR treatment with 5 μM and 10 μM compared to HULP without treatment (5 μM FC = 0.520, p-value = 0.003, 10 μM FC = 0.577, p-value = 0.007, respectively) (Fig. 4D). No significant differences were found in MMP7 expression at 2 μM (FC = 0.824, p-value = 0.860). Thus, 5-aza-CdR inhibited final targets of Wnt/β-catenin pathway in HULP cells in vitro in a dose-dependent manner.

Discussion

Our study shows that 5-aza-2′-deoxycitidine inhibits cell proliferation, ECM formation, and Wnt/β-catenin pathway in human uterine leiomyoma primary cells, suggesting DNA methyltransferases inhibitors as a new effective option for uterine leiomyoma treatment. UL is a multifactorial disease with an unclear pathogenesis and ineffective treatment [6–9]. For this reason, determining the molecular mechanisms involved in UL growth is necessary to better understand its pathogenesis, as well as define molecular targets to develop new therapeutic options against them. Factors contributing to UL pathogenesis include genetic mutation, epigenetic modifications, and several growth factors [14]. In addition, estrogen and progesterone are recognized as promoters of UL growth [14]. Accordingly, a significant growth of UL during the first trimester of pregnancy is associated with hormonal changes [41]. In this regard, epigenetic modifications could play an important role in steroid hormonal “cross-talk” for UL development and growth [14, 42]. Epigenetics is emerging as a new hallmark of tumor development with a high therapeutic application because of its potential for reversal [23, 25]. In fact, abnormal DNA methylation is found in UL compared to MM tissue, where tumor suppressor genes are hypermethylated, which contributes to the development of this tumor [14, 21, 27,28,29,30]. Based on these findings, we focused our study on DNMT methylation reversion by DNMT inhibitor 5-aza-2′-deoxycitidine as new therapeutic option for UL.

For this purpose, we first studied DNMT expression and activity of UL and MM tissue and we corroborated that there is a higher DNMT1 expression and DNMT activity in UL compared to MM tissue. Interestingly, similar findings were reported by others, who associated the aberrant DNA methylation found in UL with an increased DNMT1 and DNMT3a mRNA expression in tumor samples compared to myometrium [26, 27]. Based on these finding, the use of DNMT inhibitors to reverse the aberrant DNA methylation found in UL could be a good therapeutic approach to treat them. Secondly, since epigenetic changes depend on the environment, we studied DNMT expression and activity in cell culture to confirm that an in vitro model maintains the epigenetic feature from origin tissue. We proved for the first time that the increased DNMT activity observed in UL tissue is maintained under cell culture conditions over time in HULP compared to MM cells, confirming the reproducibility of the in vitro model. This approach allows the study of epigenetic modifications such as DNMT methylation reversion using DNMT inhibitor.

In this regard, the use of DNMT inhibitor 5-aza-CdR is widely described in several tumor types, such as colorectal, bladder, and pancreatic cancer, demonstrating antiproliferative effects [38, 43, 44]. Uncontrolled proliferation is one of the hallmarks of cancer, and tumorigenic cells present greater proliferation and lesser apoptosis than normal cells. Accordingly, several authors showed that UL growth is due to an increased proliferation of UL cells [7, 13]. Since UL is associated with aberrant DNA methylation and increased cell proliferation, reversion of DNMT methylation by 5-aza-CdR could be an efficient treatment to reduce UL size due to its antiproliferative effects. Here, we found that 5-aza-CdR treatment of HULP cells inhibited the number of proliferating cells, and this decrease was due to a reduction in cell proliferation, as demonstrated by decreased PCNA protein expression. These findings demonstrated the antiproliferative effect in UL previously described in different tumors [38, 43, 44]. Meanwhile, apoptosis did not increase after 5-aza-CdR treatment, suggesting that this treatment would not be toxic for HULP cells. Based on our results, 5-aza-CdR would decrease cell proliferation in HULP cells, highlighting its potential as a therapeutic option to reduce UL growth.

UL growth is due not only to cell proliferation, but also to an excessive synthesis and deposition of ECM. Thus, evaluating the effect of DNMT inhibitors on ECM protein expression is necessary to assess the possible use of these inhibitors as a new therapeutic option for UL. Our results showed that 5-aza-CdR treatment significantly decreased the expression of ECM-associated proteins such as FIBRONECTIN, COLLAGEN I, and PAI-1. These findings suggested an important role of 5-aza-CdR in the regulation of key fibrotic proteins involved in UL expansion. In line with this, other studies demonstrated the success of treatments targeting ECM formation as possible therapeutic option to reduce UL grown, such as Vitamin D treatment in the Eker rat model [45], in a xenograft mouse model [46], and in human leiomyoma cells [47]. Hence, 5-aza-CdR reduced ECM expansion and cell proliferation in HULP cells, which counteracts UL growth, offering a promising candidate to treat this tumor.

Wnt/β-catenin pathway is involved in several cellular functions, being a key regulator of cell proliferation and ECM production. This pathway is dysregulated in several cancers [31,32,33,34, 48] and UL [7, 13, 35]. This pathway’s implications in colorectal cancer (CRC) are well studied [49]. In CRC, the effect of 5-aza-CdR in reduction of cell self-renewal through Wnt/β-catenin pathway inhibition is demonstrated, showing the important role of epigenetic modification on Wnt/β-catenin pathway regulation and its implication in cancer development [38]. In UL, previous studies suggested that targeting Wnt/β-catenin pathway can be a promising therapeutic approach because of their aberrant activation in UL cells compared to MM cells [13, 36, 37, 50]. Due to the increased activation of Wnt/β-catenin pathway and aberrant DNA methylation found in UL, we analyzed if DNA methylation reversion by 5-aza-CdR treatment had an effect on the Wnt/β-catenin pathway in HULP cells. We assessed protein or mRNA expression of final targets of the Wnt/β-catenin pathway, such as WISP1, c-MYC, and MMP7, because of their tight association with tumor progression. We found that both c-MYC gene expression and WISP1 protein expression were downregulated in treated HULP cells. c-MYC is one of the most studied final targets of the Wnt/β-catenin pathway. It is a protooncogene involved in cell cycle progression [51] whose deregulation has been linked to an aberrant Wnt/β-catenin pathway expression in CRC [52]. WISP1 has been identified as an oncogene in several cancer types such as glioblastoma [53], CRC [54], and colon cancer [55], with involvement in tumor proliferation, migration, and poor prognosis. Inhibition of WISP1 decreases cell proliferation and invasion through increasing apoptosis and blocking cell cycle in glioblastoma and colon cancer cells [53, 55]. Therefore, the reduction of WISP1 and c-MYC expression observed in our study suggest that DNMT inhibition by 5-aza-CdR could impede cell proliferation via Wnt/β-catenin pathway inhibition. Finally, MMP7 is a metalloprotease involved in ECM physiology, epithelial-mesenchymal transition, and tumor invasion that is overexpressed in some cancer types [56]. In this regard, a study in hepatocellular carcinomas demonstrated the inhibition of cell migration and invasion after diminishing MMP7 [57]. Accordingly, we observed a decrease in MMP7 gene expression after 5-aza-CdR treatment in HULP cells, suggesting a lower invasive capacity of HULP-treated cells via Wnt/β-catenin pathway inhibition.

Based on these findings, we suggest that DNA methylation is involved in Wnt/β-catenin pathway regulation and, consequently, in cell proliferation and ECM formation in HULP cells, proposing 5-aza-CdR as a treatment to reduce uterine leiomyoma growth. Further studies are necessary to determine the role of DNA methylation on Wnt/β-catenin pathway, as well as the in vivo effect of 5-aza-CdR on UL.

Conclusions

Our study demonstrated for the first time that 5-aza-CdR reduces cell proliferation, ECM formation, and Wnt/β-catenin pathway in HULP cells in vitro. Yet, apoptosis appears unaffected. Based on our findings, we suggest DNMT inhibitors such as 5-aza-CdR could offer a new therapeutic option to treat patients with uterine leiomyoma.

Availability of data and materials

All data generated or analyzed during this study are included in this published article [and its supplementary information files].

Abbreviations

5-aza-CdR:

5-aza-2′-deoxycytidine.

AGnRH:

Gonadotropin releasing-hormone agonist

BAX:

BCL2 associated-X

BCL2:

B-cell lymphoma-2

bFGF:

Basic FGF

BMI:

Body mass index

c-MYC:

MYC proto-oncogene

COL4A5:

Collagen type IV alpha 5

COL4A6:

Collagen type IV alpha 6

COL-I :

Collagen I

CRC:

Colorectal cancer

DNMT:

DNA methyltransferase

ECM:

Extracellular matrix

FBS:

Fetal bovine serum

FGF:

Acidic fibroblast growth factor

FH:

Fumarate hydratase

GAPDH:

Glyceraldehyde-3-Phosphate Dehydrogenase

HMGA2:

High mobility group AT-hook 2

HULP:

Human uterine leiomyoma primary

MED12:

Mediator complex subunit 12

MM:

Adjacent myometrium

MMP7:

Matrix metalloproteinase 7

PAI-I:

Plasminogen activator inhibitor-1

PCNA:

Proliferating cell nuclear antigen

RIPA:

Radioimmunoprecipitation assay

TGF-β:

Tumor growth factor beta

UL:

Uterine leiomyoma

UPA:

Ulipristal acetate

VEGF:

Vascular endothelial growth factor

WB:

Western blot

WISP1:

Wnt1-inducible-signaling pathway protein 1

References

  1. Stewart EA. Uterine fibroids. Lancet. 2001;357:293–8 Elsevier Limited.

    Article  CAS  PubMed  Google Scholar 

  2. Parker WH. Etiology, symptomatology, and diagnosis of uterine myomas. Fertil Steril. 2007;87:725–36.

    Article  PubMed  Google Scholar 

  3. Marshall LM, Spiegelman D, Barbieri RL, Goldman MB, Manson JE, Colditz GA, et al. Variation in the incidence of uterine leiomyoma among premenopausal women by age and race. Obstet Gynecol. 1997;90:967–73.

    Article  CAS  PubMed  Google Scholar 

  4. 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;188:100–7 Mosby Inc.

    Article  PubMed  Google Scholar 

  5. Gupta S, Jose J, Manyonda I. Clinical presentation of fibroids. Best Pract Res Clin Obstet Gynaecol. 2008;22:615–26.

    Article  PubMed  Google Scholar 

  6. Parker JD, Malik M, Catherino WH. Human myometrium and leiomyomas express gonadotropin-releasing hormone 2 and gonadotropin-releasing hormone 2 receptor. Fertil Steril. 2007;88:39–46.

    Article  CAS  PubMed  Google Scholar 

  7. Bulun SE. Uterine fibroids. N Engl J Med. 2013;369:1344–55.

    Article  CAS  PubMed  Google Scholar 

  8. Manyonda I, Sinthamoney E, Belli AM. Controversies and challenges in the modern management of uterine fibroids. BJOG An Int J Obstet Gynaecol. 2004;111:95–102.

    Article  Google Scholar 

  9. Friedman AJ. Treatment of leiomyomata uteri with short-term leuprolide followed by leuprolide plus estrogen-progestin hormone replacement therapy for 2 years: a pilot study. Fertil Steril. 1989;51:526–8.

    Article  CAS  PubMed  Google Scholar 

  10. Maruo T, Ohara N, Wang J, Matsuo H. Sex steroidal regulation of uterine leiomyoma growth and apoptosis. Hum Reprod Update. 2004;10:207–20.

    Article  CAS  PubMed  Google Scholar 

  11. Patel B, Elguero S, Thakore S, Dahoud W, Bedaiwy M, Mesiano S. Role of nuclear progesterone receptor isoforms in uterine pathophysiology. Hum Reprod Update. 2015;21:155–73 Oxford University Press.

    Article  CAS  PubMed  Google Scholar 

  12. Sozen I, Arici A. Interactions of cytokines, growth factors, and the extracellular matrix in the cellular biology of uterine leiomyomata. Fertil Steril. 2002;78:1–12.

    Article  PubMed  Google Scholar 

  13. Ono M, Yin P, Navarro A, Moravek MB, Coon VJS, Druschitz SA, 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;110:17053–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Laganà AS, Vergara D, Favilli A, La Rosa VL, Tinelli A, Gerli S, et al. Epigenetic and genetic landscape of uterine leiomyomas: a current view over a common gynecological disease. Arch Gynecol Obstet. 2017;296:855–67 Springer Verlag.

    Article  PubMed  CAS  Google Scholar 

  15. Mehine M, Kaasinen E, Heinonen HR, Mäkinen N, Kämpjärvi K, Sarvilinna N, et al. Integrated data analysis reveals uterine leiomyoma subtypes with distinct driver pathways and biomarkers. Proc Natl Acad Sci U S A. 2016;113:1315–20 National Academy of Sciences.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Chiaffarino F, Parazzini F, La Vecchia C, Chatenoud L, Di Cintio E, Marsico S. Diet and uterine myomas. Obstet Gynecol. 1999;94:395–8 Elsevier Inc.

    CAS  PubMed  Google Scholar 

  17. Faerstein E, Szklo M, Rosenshein NB. Risk factors for uterine leiomyoma: a practice-based case-controls study. II. Atherogenic risk factors and potential sources of uterine irritation. Am J Epidemiol. 2001;153:11–9.

    Article  CAS  PubMed  Google Scholar 

  18. Faerstein E, Szklo M, Rosenshein N. Risk factors for uterine leiomyoma: a practice-based case-control study. I. African-American heritage, reproductive history, body size, and smoking. Am J Epidemiol. 2001;153:1–10.

    Article  CAS  PubMed  Google Scholar 

  19. Lopez M, Halby L, Arimondo PB. DNA methyltransferase inhibitors: development and applications. Adv Exp Med Biol. 2016;945:431–73 Springer New York LLC.

    Article  CAS  PubMed  Google Scholar 

  20. Cedar H, Bergman Y. Linking DNA methylation and histone modification: patterns and paradigms. Nat Rev Genet. 2009;10:295–304.

    Article  CAS  PubMed  Google Scholar 

  21. Yang Q, Mas A, Diamond MP, Al-Hendy A. The mechanism and function of epigenetics in uterine leiomyoma development. Reprod Sci. 2016;23:163–75 SAGE Publications Inc.

    Article  CAS  PubMed  Google Scholar 

  22. Braný D, Dvorská D, Grendár M, Ňachajová M, Szépe P, Lasabová Z, et al. Different methylation levels in the KLF4, ATF3 and DLEC1 genes in the myometrium and in corpus uteri mesenchymal tumours as assessed by MS-HRM. Pathol Res Pract. 2019;215:152465 Elsevier GmbH.

    Article  PubMed  CAS  Google Scholar 

  23. Klutstein M, Nejman D, Greenfield R, Cedar H. DNA methylation in cancer and aging. Cancer Res. 2016;76:3446–50 American Association for Cancer Research Inc.

    Article  CAS  PubMed  Google Scholar 

  24. Kobayashi Y, Absher DM, Gulzar ZG, Young SR, McKenney JK, Peehl DM, et al. DNA methylation profiling reveals novel biomarkers and important roles for DNA methyltransferases in prostate cancer. Genome Res. 2011;21:1017–27.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Yang Q, Liu S, Tian Y, Hasan C, Kersey D, Salwen HR, et al. Methylation-associated silencing of the heat shock protein 47 gene in human neuroblastoma. Cancer Res. 2004;64:4531–8.

    Article  CAS  PubMed  Google Scholar 

  26. Li S, Chiang TC, Richard-Davis G, Barrett JC, Mclachlan JA. DNA hypomethylation and imbalanced expression of DNA methyltransferases (DNMT1, 3A, and 3B) in human uterine leiomyoma. Gynecol Oncol. 2003;90:123–30 Academic Press Inc.

    Article  CAS  PubMed  Google Scholar 

  27. Yamagata Y, Maekawa R, Asada H, Taketani T, Tamura I, Tamura H, et al. Aberrant DNA methylation status in human uterine leiomyoma. Mol Hum Reprod. 2009;15:259–67.

    Article  CAS  PubMed  Google Scholar 

  28. Navarro A, Yin P, Monsivais D, Lin SM, Du P, Wei JJ, et al. Genome-wide DNA methylation indicates silencing of tumor suppressor genes in uterine leiomyoma. PLoS One. 2012;7:e33284.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Maekawa R, Sato S, Yamagata Y, Asada H, Tamura I, Lee L, et al. Genome-wide DNA methylation analysis reveals a potential mechanism for the pathogenesis and development of uterine leiomyomas. PLoS One. 2013;8:e66632.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Sato S, Maekawa R, Yamagata Y, Tamura I, Lee L, Okada M, et al. Identification of uterine leiomyoma-specific marker genes based on DNA methylation and their clinical application. Sci Rep. 2016;6:1 Nature Publishing Group.

    Article  CAS  Google Scholar 

  31. Clevers H, Nusse R. Wnt/β-catenin signaling and disease. Cell. 2012;149:1192–205.

    Article  CAS  PubMed  Google Scholar 

  32. Garg M, Maurya N. WNT/β-catenin signaling in urothelial carcinoma of bladder. World J Nephrol. 2019;8:83–94 Baishideng Publishing Group Inc.

    Article  PubMed  PubMed Central  Google Scholar 

  33. Shang S, Hua F, Hu ZW. The regulation of β-catenin activity and function in cancer: therapeutic opportunities. Oncotarget. 2017;8:33972–89 Impact Journals LLC.

    Article  PubMed  PubMed Central  Google Scholar 

  34. Zhan T, Rindtorff N, Boutros M. Wnt signaling in cancer. Oncogene. 2017;36:1461–73 Nature Publishing Group.

    Article  CAS  PubMed  Google Scholar 

  35. Borahay MA, Al Hendy A, Kilic GS, Boehning D. Signaling pathways in leiomyoma: understanding pathobiology and implications for therapy. Mol Med. 2015;21:242–56 Uninversity of Michigan.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Corachán A, Ferrero H, Aguilar A, Garcia N, Monleon J, Faus A, et al. Inhibition of tumor cell proliferation in human uterine leiomyomas by vitamin D via Wnt/β-catenin pathway. Fertil Steril. 2019;111:397–407 Elsevier Inc.

    Article  PubMed  CAS  Google Scholar 

  37. Al-Hendy A, Diamond MP, Boyer TG, Halder SK. Vitamin D3 inhibits Wnt/β-catenin and mTOR signaling pathways in human uterine fibroid cells. J Clin Endocrinol Metab. 2016;101:1542–51 Endocrine Society.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Li S, Han Z, Zhao N, Zhu B, Zhang Q, Yang X, et al. Inhibition of DNMT suppresses the stemness of colorectal cancer cells through down-regulating Wnt signaling pathway. Cell Signal. 2018;47:79–87 Elsevier Inc.

    Article  PubMed  CAS  Google Scholar 

  39. Logan PC, Ponnampalam AP, Rahnama F, Lobie PE, Mitchell MD. The effect of DNA methylation inhibitor 5-Aza-2′-deoxycytidine on human endometrial stromal cells. Hum Reprod. 2010;25:2859–69 Oxford University Press.

    Article  CAS  PubMed  Google Scholar 

  40. Bin DY, Long CL, Liu XQ, Chen XM, Guo LR, Xia YY, et al. 5-Aza-2′-deoxycytidine leads to reduced embryo implantation and reduced expression of DNA methyltransferases and essential endometrial genes. PLoS One. 2012;7:e45364.

    Article  CAS  Google Scholar 

  41. Vitagliano A, Noventa M, Di Spiezio SA, Saccone G, Gizzo S, Borgato S, et al. Uterine fibroid size modifications during pregnancy and puerperium: evidence from the first systematic review of literature. Arch Gynecol Obstet. 2018;297:823–35 Springer Verlag.

    Article  PubMed  Google Scholar 

  42. Asada H, Yamagata Y, Taketani T, Matsuoka A, Tamura H, Hattori N, et al. Potential link between estrogen receptor-α gene hypomethylation and uterine fibroid formation. Mol Hum Reprod. 2008;14:539–45.

    Article  CAS  PubMed  Google Scholar 

  43. Nunes SP, Henrique R, Jerónimo C, Paramio JM. DNA methylation as a therapeutic target for bladder cancer. Cells. 2020;9:1850 NLM (Medline).

    Article  CAS  PubMed Central  Google Scholar 

  44. Wong KK. DNMT1 as a therapeutic target in pancreatic cancer: mechanisms and clinical implications. Cell Oncol. 2020;43:779–92 Springer Science and Business Media B.V.

    Article  CAS  Google Scholar 

  45. Halder SK, Sharan C, Al-Hendy A. 1,25-Dihydroxyvitamin D3 treatment shrinks uterine leiomyoma tumors in the Eker rat model. Biol Reprod. 2012;86:116.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  46. Corachán A, Ferrero H, Escrig J, Monleon J, Faus A, Cervelló I, et al. Long-term vitamin D treatment decreases human uterine leiomyoma size in a xenograft animal model. Fertil Steril. 2020;113:205–216.e4 Elsevier Inc.

    Article  PubMed  CAS  Google Scholar 

  47. Halder SK, Osteen KG, Al-Hendy A. 1,25-Dihydroxyvitamin D3 reduces extracellular matrix-associated protein expression in human uterine fibroid cells. Biol Reprod. 2013;89:150 Society for the Study of Reproduction.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  48. Cheng Y-Y, Huang N-C, Chang Y-T, Sung J-M, Shen K-H, Tsai C-C, et al. Associations between arsenic in drinking water and the progression of chronic kidney disease: a nationwide study in Taiwan. J Hazard Mater. 2017;321:432–9 Department of Environmental and Occupational Health, College of Medical, National Cheng Kung University, Tainan, Taiwan: Elsevier.

    Article  CAS  PubMed  Google Scholar 

  49. Deitrick J, Pruitt WM. Wnt/β catenin-mediated signaling commonly altered in colorectal cancer. Prog Mol Biol Transl Sci. 2016;144:49–68 Elsevier B.V.

    Article  CAS  PubMed  Google Scholar 

  50. Ali M, Shahin SM, Sabri NA, Al-Hendy A, Yang Q. Activation of β-catenin signaling and its crosstalk with estrogen and histone deacetylases in human uterine fibroids. J Clin Endocrinol Metab. 2020;105:E1517–35 Endocrine Society.

    Article  Google Scholar 

  51. Zakiryanova GK, Wheeler S, Shurin MR. Oncogenes in immune cells as potential therapeutic targets. ImmunoTargets Ther. 2018;7:21–8 Dove Medical Press Ltd.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Rennoll S. Regulation of MYC gene expression by aberrant Wnt/β-catenin signaling in colorectal cancer. World J Biol Chem. 2015;6:290 Baishideng Publishing Group Inc.

    Article  PubMed  PubMed Central  Google Scholar 

  53. Jing D, Zhang Q, Yu H, Zhao Y, Shen L. Identification of WISP1 as a novel oncogene in glioblastoma. Int J Oncol. 2017;51:1261–70 Spandidos Publications.

    Article  CAS  PubMed  Google Scholar 

  54. Jiang. Differential expression of the CCN family member WISP-1, WISP-2 and WISP-3 in human colorectal cancer and the prognostic implications. Int J Oncol. 2010;36:1129 Spandidos Publications.

    Article  PubMed  CAS  Google Scholar 

  55. Wu J, Long Z, Cai H, Du C, Liu X, Yu S, et al. High expression of WISP1 in colon cancer is associated with apoptosis, invasion and poor prognosis. Oncotarget. 2016;7:49834–47 Impact Journals LLC.

    Article  PubMed  PubMed Central  Google Scholar 

  56. Scheau C, Badarau IA, Costache R, Caruntu C, Mihai GL, Didilescu AC, et al. The role of matrix metalloproteinases in the epithelial-mesenchymal transition of hepatocellular carcinoma. Anal Cell Pathol. 2019;2019:9423907 Hindawi Limited.

    Article  CAS  Google Scholar 

  57. Lin Y, Liu J, Huang Y, Liu D, Zhang G, Kan H. microRNA-489 plays an anti-metastatic role in human hepatocellular carcinoma by targeting matrix metalloproteinase-7. Transl Oncol. 2017;10:211–20 Neoplasia Press, Inc.

    Article  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

The authors express sincere thanks to participants of this study who made this work possible and to all medical staff of the Hospital Universitario y Politécnico La Fe, Valencia for their assistance in obtaining samples.

Funding

This study was supported by Health Institute Carlos III, Fondo Europeo de Desarrollo Regional (FEDER) awarded to A.P. (PI18/00323) and I.C. (CP19/00149) and by Miguel Servet Program awarded to H.F. (CP20/00120), by Generalitat Valenciana awarded to A.P. (PROMETEO/2018/137) and VALi+d Programe awarded to M.C.C.-G. (ACIF/2019/139) and to A.C. (APOSTD/2020/123), as well as by the Spanish Ministry of Education through FPU to M.S. (FPU18/03735).

Author information

Authors and Affiliations

Authors

Contributions

M.C.C.-G. was involved in study design, executed experiments, and wrote and edited the manuscript. A.C. was involved in experimental execution and wrote the manuscript. M.S. analyzed results and executed QT-RT-PCR to study gene expression. J. E and J.M. were involved in selection and recruitment of UL patients and sample collection by processing UL and MM biopsies at surgery. A.F. was involved in sample collection by participating in HULP cell isolation. A.P. devised and supervised the study, contributed to data interpretation, and drafted the manuscript. I.C. and H. F coordinated the study design, contributed to data interpretation, and edited the manuscript. All authors reviewed the manuscript and provided critical feedback and discussion. The author(s) read and approved the final manuscript.

Corresponding author

Correspondence to Hortensia Ferrero.

Ethics declarations

Ethics approval and consent to participate

This study was approved by the Clinical Ethics Committee at the Hospital Univeristario y Politecnico La Fe (Spain) (2018/0097), and all participants provided informed consent.

Consent for publication

Yes.

Competing interests

The authors have no conflicts of interest to declare.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary Information

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Carbajo-García, M.C., Corachán, A., Segura-Benitez, M. et al. 5-aza-2′-deoxycitidine inhibits cell proliferation, extracellular matrix formation and Wnt/β-catenin pathway in human uterine leiomyomas. Reprod Biol Endocrinol 19, 106 (2021). https://doi.org/10.1186/s12958-021-00790-5

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12958-021-00790-5

Keywords