Skip to main content
Top
Published in: Reproductive Biology and Endocrinology 1/2021

Open Access 01-12-2021 | Epigenetics | Research

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

Authors: María Cristina Carbajo-García, Ana Corachán, Marina Segura-Benitez, Javier Monleón, Julia Escrig, Amparo Faus, Antonio Pellicer, Irene Cervelló, Hortensia Ferrero

Published in: Reproductive Biology and Endocrinology | Issue 1/2021

Login to get access

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.
Appendix
Available only for authorised users
Literature
2.
go back to reference Parker WH. Etiology, symptomatology, and diagnosis of uterine myomas. Fertil Steril. 2007;87:725–36.PubMedCrossRef Parker WH. Etiology, symptomatology, and diagnosis of uterine myomas. Fertil Steril. 2007;87:725–36.PubMedCrossRef
3.
go back to reference 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.PubMedCrossRef 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.PubMedCrossRef
4.
go back to reference 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.PubMedCrossRef 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.PubMedCrossRef
5.
go back to reference Gupta S, Jose J, Manyonda I. Clinical presentation of fibroids. Best Pract Res Clin Obstet Gynaecol. 2008;22:615–26.PubMedCrossRef Gupta S, Jose J, Manyonda I. Clinical presentation of fibroids. Best Pract Res Clin Obstet Gynaecol. 2008;22:615–26.PubMedCrossRef
6.
go back to reference 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.PubMedCrossRef 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.PubMedCrossRef
8.
go back to reference 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.CrossRef 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.CrossRef
9.
go back to reference 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.PubMedCrossRef 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.PubMedCrossRef
10.
go back to reference Maruo T, Ohara N, Wang J, Matsuo H. Sex steroidal regulation of uterine leiomyoma growth and apoptosis. Hum Reprod Update. 2004;10:207–20.PubMedCrossRef Maruo T, Ohara N, Wang J, Matsuo H. Sex steroidal regulation of uterine leiomyoma growth and apoptosis. Hum Reprod Update. 2004;10:207–20.PubMedCrossRef
11.
go back to reference 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.PubMedCrossRef 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.PubMedCrossRef
12.
go back to reference 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.PubMedCrossRef 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.PubMedCrossRef
13.
go back to reference 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.PubMedPubMedCentralCrossRef 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.PubMedPubMedCentralCrossRef
14.
go back to reference 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.PubMedCrossRef 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.PubMedCrossRef
15.
go back to reference 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.PubMedPubMedCentralCrossRef 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.PubMedPubMedCentralCrossRef
16.
go back to reference 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.PubMed 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.PubMed
17.
go back to reference 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.PubMedCrossRef 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.PubMedCrossRef
18.
go back to reference 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.PubMedCrossRef 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.PubMedCrossRef
19.
go back to reference Lopez M, Halby L, Arimondo PB. DNA methyltransferase inhibitors: development and applications. Adv Exp Med Biol. 2016;945:431–73 Springer New York LLC.PubMedCrossRef Lopez M, Halby L, Arimondo PB. DNA methyltransferase inhibitors: development and applications. Adv Exp Med Biol. 2016;945:431–73 Springer New York LLC.PubMedCrossRef
20.
go back to reference Cedar H, Bergman Y. Linking DNA methylation and histone modification: patterns and paradigms. Nat Rev Genet. 2009;10:295–304.CrossRefPubMed Cedar H, Bergman Y. Linking DNA methylation and histone modification: patterns and paradigms. Nat Rev Genet. 2009;10:295–304.CrossRefPubMed
21.
go back to reference 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.PubMedCrossRef 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.PubMedCrossRef
22.
go back to reference 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.PubMedCrossRef 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.PubMedCrossRef
23.
go back to reference 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.PubMedCrossRef 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.PubMedCrossRef
24.
go back to reference 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.PubMedPubMedCentralCrossRef 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.PubMedPubMedCentralCrossRef
25.
go back to reference 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.PubMedCrossRef 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.PubMedCrossRef
26.
go back to reference 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.PubMedCrossRef 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.PubMedCrossRef
27.
go back to reference 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.PubMedCrossRef 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.PubMedCrossRef
28.
go back to reference 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.PubMedPubMedCentralCrossRef 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.PubMedPubMedCentralCrossRef
29.
go back to reference 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.PubMedPubMedCentralCrossRef 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.PubMedPubMedCentralCrossRef
30.
go back to reference 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.CrossRef 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.CrossRef
32.
33.
go back to reference 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.PubMedPubMedCentralCrossRef 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.PubMedPubMedCentralCrossRef
34.
go back to reference Zhan T, Rindtorff N, Boutros M. Wnt signaling in cancer. Oncogene. 2017;36:1461–73 Nature Publishing Group.PubMedCrossRef Zhan T, Rindtorff N, Boutros M. Wnt signaling in cancer. Oncogene. 2017;36:1461–73 Nature Publishing Group.PubMedCrossRef
35.
go back to reference 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.PubMedPubMedCentralCrossRef 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.PubMedPubMedCentralCrossRef
36.
go back to reference 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.PubMedCrossRef 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.PubMedCrossRef
37.
go back to reference 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.PubMedPubMedCentralCrossRef 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.PubMedPubMedCentralCrossRef
38.
go back to reference 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.PubMedCrossRef 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.PubMedCrossRef
39.
go back to reference 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.PubMedCrossRef 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.PubMedCrossRef
40.
go back to reference 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.CrossRef 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.CrossRef
41.
go back to reference 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.PubMedCrossRef 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.PubMedCrossRef
42.
go back to reference 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.PubMedCrossRef 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.PubMedCrossRef
43.
go back to reference Nunes SP, Henrique R, Jerónimo C, Paramio JM. DNA methylation as a therapeutic target for bladder cancer. Cells. 2020;9:1850 NLM (Medline).PubMedCentralCrossRef Nunes SP, Henrique R, Jerónimo C, Paramio JM. DNA methylation as a therapeutic target for bladder cancer. Cells. 2020;9:1850 NLM (Medline).PubMedCentralCrossRef
44.
go back to reference 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.CrossRef 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.CrossRef
45.
46.
go back to reference 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.PubMedCrossRef 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.PubMedCrossRef
47.
go back to reference 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.PubMedPubMedCentralCrossRef 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.PubMedPubMedCentralCrossRef
48.
go back to reference 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.PubMedCrossRef 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.PubMedCrossRef
49.
go back to reference 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.PubMedCrossRef 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.PubMedCrossRef
50.
go back to reference 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.CrossRef 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.CrossRef
51.
go back to reference Zakiryanova GK, Wheeler S, Shurin MR. Oncogenes in immune cells as potential therapeutic targets. ImmunoTargets Ther. 2018;7:21–8 Dove Medical Press Ltd.PubMedPubMedCentralCrossRef Zakiryanova GK, Wheeler S, Shurin MR. Oncogenes in immune cells as potential therapeutic targets. ImmunoTargets Ther. 2018;7:21–8 Dove Medical Press Ltd.PubMedPubMedCentralCrossRef
52.
go back to reference 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.PubMedPubMedCentralCrossRef 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.PubMedPubMedCentralCrossRef
53.
go back to reference 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.PubMedCrossRef 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.PubMedCrossRef
54.
go back to reference 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.PubMedCrossRef 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.PubMedCrossRef
55.
go back to reference 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.PubMedPubMedCentralCrossRef 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.PubMedPubMedCentralCrossRef
56.
go back to reference 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.CrossRef 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.CrossRef
57.
go back to reference 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.PubMedPubMedCentralCrossRef 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.PubMedPubMedCentralCrossRef
Metadata
Title
5-aza-2′-deoxycitidine inhibits cell proliferation, extracellular matrix formation and Wnt/β-catenin pathway in human uterine leiomyomas
Authors
María Cristina Carbajo-García
Ana Corachán
Marina Segura-Benitez
Javier Monleón
Julia Escrig
Amparo Faus
Antonio Pellicer
Irene Cervelló
Hortensia Ferrero
Publication date
01-12-2021
Publisher
BioMed Central
Keyword
Epigenetics
Published in
Reproductive Biology and Endocrinology / Issue 1/2021
Electronic ISSN: 1477-7827
DOI
https://doi.org/10.1186/s12958-021-00790-5

Other articles of this Issue 1/2021

Reproductive Biology and Endocrinology 1/2021 Go to the issue