Human umbilical cord blood-derived MSCs trans-differentiate into endometrial cells and regulate Th17/Treg balance through NF-κB signaling in rabbit intrauterine adhesions endometrium

Abstract Purpose The fundamental cause of intrauterine adhesions (IUAs) is the destruction and reduction in stem cells in endometrial basal layer, resulting in endometrial reconstruction very difficult. The purpose of this study was to investigate the effects and underlying mechanism of human umbili...

Full description

Bibliographic Details
Main Authors: Qing Hua, Yong Zhang, Hongjuan Li, Haoran Li, Ranran Jin, Li Li, Yuancui Xiang, Meng Tian, Jingjing Wang, Lei Sun, Yali Wang
Format: Article
Language:English
Published: BMC 2022-07-01
Series:Stem Cell Research & Therapy
Subjects:
Online Access:https://doi.org/10.1186/s13287-022-02990-1
_version_ 1818197099530944512
author Qing Hua
Yong Zhang
Hongjuan Li
Haoran Li
Ranran Jin
Li Li
Yuancui Xiang
Meng Tian
Jingjing Wang
Lei Sun
Yali Wang
author_facet Qing Hua
Yong Zhang
Hongjuan Li
Haoran Li
Ranran Jin
Li Li
Yuancui Xiang
Meng Tian
Jingjing Wang
Lei Sun
Yali Wang
author_sort Qing Hua
collection DOAJ
description Abstract Purpose The fundamental cause of intrauterine adhesions (IUAs) is the destruction and reduction in stem cells in endometrial basal layer, resulting in endometrial reconstruction very difficult. The purpose of this study was to investigate the effects and underlying mechanism of human umbilical cord blood-derived mesenchymal stem cells (hUCB-MSCs) on the endometrial reconstruction after transplantation. Methods hUCB-MSCs were isolated and identified by flow cytometry, osteogenic, adipogenic and chondrogenic differentiation assays. The rabbit IUA models were established and set five groups (control, 14/28th day after surgery, estrogen and hUCB-MSCs treatment). The number of endometrial glands and the fibrosis rate were evaluated using HE and Masson staining, respectively. Endometrial proliferation, angiogenesis and inflammation were evaluated by immunohistochemical staining of ER, Ki-67and TGF-β1, respectively. Single-cell RNA sequencing (scRNA-seq) was applied to explore the cell differentiation trajectory after hUCB-MSCs transplanted into IUA endometrium. Finally, molecular mechanism of hUCB-MSCs repairing damaged endometrium was investigated by RNA sequencing, qRT-PCR and Western blot assays. Results After transplantation of the hUCB-MSCs, the increase in endometrial gland number, estrogen receptor (ER) and Ki-67 expression, and the decrease in fibrosis rate and TGF-β expression (P < 0.05), suggested the endometrial repair, angiogenesis and inflammatory suppression. The therapeutic effect of hUCB-MSCs was significantly improved compared with 28th day after surgery and estrogen group. ScRNA-seq demonstrated that the transplanted hUCB-MSCs can trans-differentiate into endometrial cells: epithelial, fibroblast and macrophage. RNA sequencing of six IUA samples combined with qRT-PCR and Western blot assays further revealed that hUCB-MSCs may regulate Th17/Treg balance through NF-κB signaling, thus inhibiting the immune response of damaged endometrium. Conclusions Our study demonstrated that hUCB-MSCs can repair damaged endometrium through trans-differentiation, immunomodulatory capacities and NF-κB signaling, suggesting the treatment value of hUCB-MSCs in IUA.
first_indexed 2024-12-12T01:44:35Z
format Article
id doaj.art-8e642e0ab2514c89b7e0cb2fe08f7ff2
institution Directory Open Access Journal
issn 1757-6512
language English
last_indexed 2024-12-12T01:44:35Z
publishDate 2022-07-01
publisher BMC
record_format Article
series Stem Cell Research & Therapy
spelling doaj.art-8e642e0ab2514c89b7e0cb2fe08f7ff22022-12-22T00:42:37ZengBMCStem Cell Research & Therapy1757-65122022-07-0113111510.1186/s13287-022-02990-1Human umbilical cord blood-derived MSCs trans-differentiate into endometrial cells and regulate Th17/Treg balance through NF-κB signaling in rabbit intrauterine adhesions endometriumQing Hua0Yong Zhang1Hongjuan Li2Haoran Li3Ranran Jin4Li Li5Yuancui Xiang6Meng Tian7Jingjing Wang8Lei Sun9Yali Wang10Department of Obstetrics and Gynecology, Zhengzhou Central Hospital Affiliated to Zhengzhou UniversityBranch Center of Advanced Medical Research Center, Zhengzhou Central Hospital Affiliated to Zhengzhou UniversityDepartment of Obstetrics and Gynecology, Zhengzhou Central Hospital Affiliated to Zhengzhou UniversityDepartment of Obstetrics and Gynecology, Zhengzhou Central Hospital Affiliated to Zhengzhou UniversityDepartment of Translational Medicine Center, Zhengzhou Central Hospital Affiliated to Zhengzhou UniversityDepartment of Obstetrics and Gynecology, Zhengzhou Central Hospital Affiliated to Zhengzhou UniversityDepartment of Obstetrics and Gynecology, Zhengzhou Central Hospital Affiliated to Zhengzhou UniversityDepartment of Obstetrics and Gynecology, Zhengzhou Central Hospital Affiliated to Zhengzhou UniversityDepartment of Obstetrics and Gynecology, Zhengzhou Central Hospital Affiliated to Zhengzhou UniversityDepartment of Obstetrics and Gynecology, Zhengzhou Central Hospital Affiliated to Zhengzhou UniversityDepartment of Obstetrics and Gynecology, Zhengzhou Central Hospital Affiliated to Zhengzhou UniversityAbstract Purpose The fundamental cause of intrauterine adhesions (IUAs) is the destruction and reduction in stem cells in endometrial basal layer, resulting in endometrial reconstruction very difficult. The purpose of this study was to investigate the effects and underlying mechanism of human umbilical cord blood-derived mesenchymal stem cells (hUCB-MSCs) on the endometrial reconstruction after transplantation. Methods hUCB-MSCs were isolated and identified by flow cytometry, osteogenic, adipogenic and chondrogenic differentiation assays. The rabbit IUA models were established and set five groups (control, 14/28th day after surgery, estrogen and hUCB-MSCs treatment). The number of endometrial glands and the fibrosis rate were evaluated using HE and Masson staining, respectively. Endometrial proliferation, angiogenesis and inflammation were evaluated by immunohistochemical staining of ER, Ki-67and TGF-β1, respectively. Single-cell RNA sequencing (scRNA-seq) was applied to explore the cell differentiation trajectory after hUCB-MSCs transplanted into IUA endometrium. Finally, molecular mechanism of hUCB-MSCs repairing damaged endometrium was investigated by RNA sequencing, qRT-PCR and Western blot assays. Results After transplantation of the hUCB-MSCs, the increase in endometrial gland number, estrogen receptor (ER) and Ki-67 expression, and the decrease in fibrosis rate and TGF-β expression (P < 0.05), suggested the endometrial repair, angiogenesis and inflammatory suppression. The therapeutic effect of hUCB-MSCs was significantly improved compared with 28th day after surgery and estrogen group. ScRNA-seq demonstrated that the transplanted hUCB-MSCs can trans-differentiate into endometrial cells: epithelial, fibroblast and macrophage. RNA sequencing of six IUA samples combined with qRT-PCR and Western blot assays further revealed that hUCB-MSCs may regulate Th17/Treg balance through NF-κB signaling, thus inhibiting the immune response of damaged endometrium. Conclusions Our study demonstrated that hUCB-MSCs can repair damaged endometrium through trans-differentiation, immunomodulatory capacities and NF-κB signaling, suggesting the treatment value of hUCB-MSCs in IUA.https://doi.org/10.1186/s13287-022-02990-1Intrauterine adhesion (IUA)Human umbilical cord blood-derived mesenchymal stem cells (hUCB-MSCs)scRNA-seqTrans-differentiationNF-κB signaling
spellingShingle Qing Hua
Yong Zhang
Hongjuan Li
Haoran Li
Ranran Jin
Li Li
Yuancui Xiang
Meng Tian
Jingjing Wang
Lei Sun
Yali Wang
Human umbilical cord blood-derived MSCs trans-differentiate into endometrial cells and regulate Th17/Treg balance through NF-κB signaling in rabbit intrauterine adhesions endometrium
Stem Cell Research & Therapy
Intrauterine adhesion (IUA)
Human umbilical cord blood-derived mesenchymal stem cells (hUCB-MSCs)
scRNA-seq
Trans-differentiation
NF-κB signaling
title Human umbilical cord blood-derived MSCs trans-differentiate into endometrial cells and regulate Th17/Treg balance through NF-κB signaling in rabbit intrauterine adhesions endometrium
title_full Human umbilical cord blood-derived MSCs trans-differentiate into endometrial cells and regulate Th17/Treg balance through NF-κB signaling in rabbit intrauterine adhesions endometrium
title_fullStr Human umbilical cord blood-derived MSCs trans-differentiate into endometrial cells and regulate Th17/Treg balance through NF-κB signaling in rabbit intrauterine adhesions endometrium
title_full_unstemmed Human umbilical cord blood-derived MSCs trans-differentiate into endometrial cells and regulate Th17/Treg balance through NF-κB signaling in rabbit intrauterine adhesions endometrium
title_short Human umbilical cord blood-derived MSCs trans-differentiate into endometrial cells and regulate Th17/Treg balance through NF-κB signaling in rabbit intrauterine adhesions endometrium
title_sort human umbilical cord blood derived mscs trans differentiate into endometrial cells and regulate th17 treg balance through nf κb signaling in rabbit intrauterine adhesions endometrium
topic Intrauterine adhesion (IUA)
Human umbilical cord blood-derived mesenchymal stem cells (hUCB-MSCs)
scRNA-seq
Trans-differentiation
NF-κB signaling
url https://doi.org/10.1186/s13287-022-02990-1
work_keys_str_mv AT qinghua humanumbilicalcordbloodderivedmscstransdifferentiateintoendometrialcellsandregulateth17tregbalancethroughnfkbsignalinginrabbitintrauterineadhesionsendometrium
AT yongzhang humanumbilicalcordbloodderivedmscstransdifferentiateintoendometrialcellsandregulateth17tregbalancethroughnfkbsignalinginrabbitintrauterineadhesionsendometrium
AT hongjuanli humanumbilicalcordbloodderivedmscstransdifferentiateintoendometrialcellsandregulateth17tregbalancethroughnfkbsignalinginrabbitintrauterineadhesionsendometrium
AT haoranli humanumbilicalcordbloodderivedmscstransdifferentiateintoendometrialcellsandregulateth17tregbalancethroughnfkbsignalinginrabbitintrauterineadhesionsendometrium
AT ranranjin humanumbilicalcordbloodderivedmscstransdifferentiateintoendometrialcellsandregulateth17tregbalancethroughnfkbsignalinginrabbitintrauterineadhesionsendometrium
AT lili humanumbilicalcordbloodderivedmscstransdifferentiateintoendometrialcellsandregulateth17tregbalancethroughnfkbsignalinginrabbitintrauterineadhesionsendometrium
AT yuancuixiang humanumbilicalcordbloodderivedmscstransdifferentiateintoendometrialcellsandregulateth17tregbalancethroughnfkbsignalinginrabbitintrauterineadhesionsendometrium
AT mengtian humanumbilicalcordbloodderivedmscstransdifferentiateintoendometrialcellsandregulateth17tregbalancethroughnfkbsignalinginrabbitintrauterineadhesionsendometrium
AT jingjingwang humanumbilicalcordbloodderivedmscstransdifferentiateintoendometrialcellsandregulateth17tregbalancethroughnfkbsignalinginrabbitintrauterineadhesionsendometrium
AT leisun humanumbilicalcordbloodderivedmscstransdifferentiateintoendometrialcellsandregulateth17tregbalancethroughnfkbsignalinginrabbitintrauterineadhesionsendometrium
AT yaliwang humanumbilicalcordbloodderivedmscstransdifferentiateintoendometrialcellsandregulateth17tregbalancethroughnfkbsignalinginrabbitintrauterineadhesionsendometrium