Cargando…

Human amniotic epithelial cells improve fertility in an intrauterine adhesion mouse model

BACKGROUND: Intrauterine adhesion (IUA) is an adhesion of the uterine cavity or cervical canal resulting from damage to the basal layer of the endometrium; this condition is usually accompanied by fibrosis of the endometrium. Previous studies have demonstrated that human amniotic epithelial cells (h...

Descripción completa

Detalles Bibliográficos
Autores principales: Li, Boning, Zhang, Qiuwan, Sun, Junyan, Lai, Dongmei
Formato: Online Artículo Texto
Lenguaje:English
Publicado: BioMed Central 2019
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6694540/
https://www.ncbi.nlm.nih.gov/pubmed/31412924
http://dx.doi.org/10.1186/s13287-019-1368-9
_version_ 1783443844954587136
author Li, Boning
Zhang, Qiuwan
Sun, Junyan
Lai, Dongmei
author_facet Li, Boning
Zhang, Qiuwan
Sun, Junyan
Lai, Dongmei
author_sort Li, Boning
collection PubMed
description BACKGROUND: Intrauterine adhesion (IUA) is an adhesion of the uterine cavity or cervical canal resulting from damage to the basal layer of the endometrium; this condition is usually accompanied by fibrosis of the endometrium. Previous studies have demonstrated that human amniotic epithelial cells (hAECs) have stem cell characteristics; however, it is unclear whether hAECs have the therapeutic potential to restore fertility after IUA. METHODS: A murine IUA model was established by mechanical injury to the uterus. Then, 10(6) hAECs were transplanted by intraperitoneal injection. The endometrium thickness, number of glands, and fibrosis area were measured by hematoxylin and eosin (H&E) staining and Masson staining. Molecules (including vWF, VEGF, PCNA, ER, PR, LC3, and p62) related to endometrial angiogenesis, cell proliferation, and autophagy were assayed by IHC staining. Pregnancy outcomes were also evaluated. Finally, hAECs were cocultured with human endometrial mesenchymal stem cells (hEnSCs) damaged by H(2)O(2) to verify the paracrine effect on endometrial stromal cells in vitro. RESULTS: The IUA uterine cavity presented with adhesion and even atresia, accompanied by a thinner endometrium, fewer glands, increased fibrosis area, and fewer microvessels. However, hAECs significantly improved the uterine structure after IUA. After hAEC treatment, the endometrium was thicker, the number of endometrial glands was increased, fibrosis was reduced, and more microvessels were generated. The expression levels of VEGF, PCNA, and ER were increased in the hAEC-treated endometrium, indicating improvements in angiogenesis and stromal cell proliferation. hAECs also increased pregnancy outcomes in IUA mice, and the pregnancy rate and fetus number increased. Furthermore, we observed altered autophagy in the IUA uterine model, and hAEC transplantation upregulated autophagy. An in vitro study showed that hAECs activated autophagy in (hEnSCs) treated with H(2)O(2) in a paracrine manner. CONCLUSIONS: Our results demonstrated that hAECs have the potential to repair the uterus after injury, providing a new strategy for the prevention and treatment of Asherman syndrome.
format Online
Article
Text
id pubmed-6694540
institution National Center for Biotechnology Information
language English
publishDate 2019
publisher BioMed Central
record_format MEDLINE/PubMed
spelling pubmed-66945402019-08-19 Human amniotic epithelial cells improve fertility in an intrauterine adhesion mouse model Li, Boning Zhang, Qiuwan Sun, Junyan Lai, Dongmei Stem Cell Res Ther Research BACKGROUND: Intrauterine adhesion (IUA) is an adhesion of the uterine cavity or cervical canal resulting from damage to the basal layer of the endometrium; this condition is usually accompanied by fibrosis of the endometrium. Previous studies have demonstrated that human amniotic epithelial cells (hAECs) have stem cell characteristics; however, it is unclear whether hAECs have the therapeutic potential to restore fertility after IUA. METHODS: A murine IUA model was established by mechanical injury to the uterus. Then, 10(6) hAECs were transplanted by intraperitoneal injection. The endometrium thickness, number of glands, and fibrosis area were measured by hematoxylin and eosin (H&E) staining and Masson staining. Molecules (including vWF, VEGF, PCNA, ER, PR, LC3, and p62) related to endometrial angiogenesis, cell proliferation, and autophagy were assayed by IHC staining. Pregnancy outcomes were also evaluated. Finally, hAECs were cocultured with human endometrial mesenchymal stem cells (hEnSCs) damaged by H(2)O(2) to verify the paracrine effect on endometrial stromal cells in vitro. RESULTS: The IUA uterine cavity presented with adhesion and even atresia, accompanied by a thinner endometrium, fewer glands, increased fibrosis area, and fewer microvessels. However, hAECs significantly improved the uterine structure after IUA. After hAEC treatment, the endometrium was thicker, the number of endometrial glands was increased, fibrosis was reduced, and more microvessels were generated. The expression levels of VEGF, PCNA, and ER were increased in the hAEC-treated endometrium, indicating improvements in angiogenesis and stromal cell proliferation. hAECs also increased pregnancy outcomes in IUA mice, and the pregnancy rate and fetus number increased. Furthermore, we observed altered autophagy in the IUA uterine model, and hAEC transplantation upregulated autophagy. An in vitro study showed that hAECs activated autophagy in (hEnSCs) treated with H(2)O(2) in a paracrine manner. CONCLUSIONS: Our results demonstrated that hAECs have the potential to repair the uterus after injury, providing a new strategy for the prevention and treatment of Asherman syndrome. BioMed Central 2019-08-14 /pmc/articles/PMC6694540/ /pubmed/31412924 http://dx.doi.org/10.1186/s13287-019-1368-9 Text en © The Author(s). 2019 Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. 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.
spellingShingle Research
Li, Boning
Zhang, Qiuwan
Sun, Junyan
Lai, Dongmei
Human amniotic epithelial cells improve fertility in an intrauterine adhesion mouse model
title Human amniotic epithelial cells improve fertility in an intrauterine adhesion mouse model
title_full Human amniotic epithelial cells improve fertility in an intrauterine adhesion mouse model
title_fullStr Human amniotic epithelial cells improve fertility in an intrauterine adhesion mouse model
title_full_unstemmed Human amniotic epithelial cells improve fertility in an intrauterine adhesion mouse model
title_short Human amniotic epithelial cells improve fertility in an intrauterine adhesion mouse model
title_sort human amniotic epithelial cells improve fertility in an intrauterine adhesion mouse model
topic Research
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6694540/
https://www.ncbi.nlm.nih.gov/pubmed/31412924
http://dx.doi.org/10.1186/s13287-019-1368-9
work_keys_str_mv AT liboning humanamnioticepithelialcellsimprovefertilityinanintrauterineadhesionmousemodel
AT zhangqiuwan humanamnioticepithelialcellsimprovefertilityinanintrauterineadhesionmousemodel
AT sunjunyan humanamnioticepithelialcellsimprovefertilityinanintrauterineadhesionmousemodel
AT laidongmei humanamnioticepithelialcellsimprovefertilityinanintrauterineadhesionmousemodel