si-SNHG5-FOXF2 inhibits TGF-β1-induced fibrosis in human primary endometrial stromal cells by the Wnt/β-catenin signalling pathway

Abstract Background Intrauterine adhesions (IUAs) are manifestations of endometrial fibrosis characterized by inflammation and fibrinogen aggregation in the extracellular matrix (ECM). The available therapeutic interventions for IUA are insufficiently effective in the clinical setting for postoperat...

Full description

Bibliographic Details
Main Authors: Limin Liu, Guobin Chen, Taoliang Chen, Wenjuan Shi, Haiyan Hu, Kaijing Song, Ruichun Huang, Huihua Cai, Yuanli He
Format: Article
Language:English
Published: BMC 2020-11-01
Series:Stem Cell Research & Therapy
Subjects:
IUA
ECM
Online Access:http://link.springer.com/article/10.1186/s13287-020-01990-3
id doaj-88aba6a5bb38493585cf2b1d19ba9e7a
record_format Article
collection DOAJ
language English
format Article
sources DOAJ
author Limin Liu
Guobin Chen
Taoliang Chen
Wenjuan Shi
Haiyan Hu
Kaijing Song
Ruichun Huang
Huihua Cai
Yuanli He
spellingShingle Limin Liu
Guobin Chen
Taoliang Chen
Wenjuan Shi
Haiyan Hu
Kaijing Song
Ruichun Huang
Huihua Cai
Yuanli He
si-SNHG5-FOXF2 inhibits TGF-β1-induced fibrosis in human primary endometrial stromal cells by the Wnt/β-catenin signalling pathway
Stem Cell Research & Therapy
IUA
TGF-β1
FOXF2
SNHG5
ECM
Fibrosis
author_facet Limin Liu
Guobin Chen
Taoliang Chen
Wenjuan Shi
Haiyan Hu
Kaijing Song
Ruichun Huang
Huihua Cai
Yuanli He
author_sort Limin Liu
title si-SNHG5-FOXF2 inhibits TGF-β1-induced fibrosis in human primary endometrial stromal cells by the Wnt/β-catenin signalling pathway
title_short si-SNHG5-FOXF2 inhibits TGF-β1-induced fibrosis in human primary endometrial stromal cells by the Wnt/β-catenin signalling pathway
title_full si-SNHG5-FOXF2 inhibits TGF-β1-induced fibrosis in human primary endometrial stromal cells by the Wnt/β-catenin signalling pathway
title_fullStr si-SNHG5-FOXF2 inhibits TGF-β1-induced fibrosis in human primary endometrial stromal cells by the Wnt/β-catenin signalling pathway
title_full_unstemmed si-SNHG5-FOXF2 inhibits TGF-β1-induced fibrosis in human primary endometrial stromal cells by the Wnt/β-catenin signalling pathway
title_sort si-snhg5-foxf2 inhibits tgf-β1-induced fibrosis in human primary endometrial stromal cells by the wnt/β-catenin signalling pathway
publisher BMC
series Stem Cell Research & Therapy
issn 1757-6512
publishDate 2020-11-01
description Abstract Background Intrauterine adhesions (IUAs) are manifestations of endometrial fibrosis characterized by inflammation and fibrinogen aggregation in the extracellular matrix (ECM). The available therapeutic interventions for IUA are insufficiently effective in the clinical setting for postoperative adhesion recurrence and infertility problems. In this study, we investigated whether si-SNHG5-FOXF2 can serve as a molecular mechanism for the inhibition of IUA fibrosis ex vivo. Methods FOXF2, TGF-β1 and collagen expression levels were measured by microarray sequencing analysis in three normal endometrium groups and six IUA patients. We induced primary human endometrial stromal cells (HESCs) into myofibroblasts (MFs) to develop an IUA cell model with various concentrations of TGF-β1 at various times. Downstream target genes of FOXF2 were screened by chromatin immunoprecipitation combined with whole-genome high-throughput sequencing (ChIP-seq). We investigated ECM formation, cell proliferation and Wnt/β-catenin signalling pathway-related proteins in primary HESCs with FOXF2 downregulation by quantitative reverse transcription-polymerase chain reaction (qRT-PCR), western blotting (WB), immunohistochemistry (IHC), flow cytometry, ethylenediurea (EdU) and CCK8 assays. We identified long noncoding RNAs (lncRNA) SNHG5 as the upstream regulatory gene of FOXF2 through RNA immunoprecipitation (RIP), RNA pulldown and fluorescence in situ hybridization (FISH). Finally, we examined FOXF2 expression, ECM formation, cell proliferation and Wnt/β-catenin signalling pathway-related proteins in primary HESCs upon FOXF2 downregulation. Results FOXF2 was highly expressed in the endometrium of patients with IUA. Treatment of primary HESCs with 10 ng/ml TGF-β1 for 72 h was found to be most effective for developing an IUA cell model. FOXF2 regulated multiple downstream target genes, including collagen, vimentin (VIM) and cyclin D2/DK4, by ChIP-seq and ChIP-PCR. FOXF2 downregulation inhibited TGF-β1-mediated primary HESC fibrosis, including ECM formation, cell proliferation and Wnt/β-catenin signalling pathway-related protein expression. We identified lncRNA SNHG5 as an upstream gene that directly regulates FOXF2 by RIP-seq, qRT-PCR, WB and FISH. SNHG5 downregulation suppressed FOXF2 expression in the IUA cell model, resulting in synergistic repression of the Wnt/β-catenin pathway, thereby altering TGF-β1-mediated ECM aggregation in endometrial stromal cells ex vivo. Conclusions Regulation of the Wnt/β-catenin signalling pathway and ECM formation by si-SNHG5-FOXF2 effectively inhibited the profibrotic effect of TGF-β1 on primary HESCs. This finding can provide a molecular basis for antagonizing TGF-β1-mediated fibrosis in primary HESCs.
topic IUA
TGF-β1
FOXF2
SNHG5
ECM
Fibrosis
url http://link.springer.com/article/10.1186/s13287-020-01990-3
work_keys_str_mv AT liminliu sisnhg5foxf2inhibitstgfb1inducedfibrosisinhumanprimaryendometrialstromalcellsbythewntbcateninsignallingpathway
AT guobinchen sisnhg5foxf2inhibitstgfb1inducedfibrosisinhumanprimaryendometrialstromalcellsbythewntbcateninsignallingpathway
AT taoliangchen sisnhg5foxf2inhibitstgfb1inducedfibrosisinhumanprimaryendometrialstromalcellsbythewntbcateninsignallingpathway
AT wenjuanshi sisnhg5foxf2inhibitstgfb1inducedfibrosisinhumanprimaryendometrialstromalcellsbythewntbcateninsignallingpathway
AT haiyanhu sisnhg5foxf2inhibitstgfb1inducedfibrosisinhumanprimaryendometrialstromalcellsbythewntbcateninsignallingpathway
AT kaijingsong sisnhg5foxf2inhibitstgfb1inducedfibrosisinhumanprimaryendometrialstromalcellsbythewntbcateninsignallingpathway
AT ruichunhuang sisnhg5foxf2inhibitstgfb1inducedfibrosisinhumanprimaryendometrialstromalcellsbythewntbcateninsignallingpathway
AT huihuacai sisnhg5foxf2inhibitstgfb1inducedfibrosisinhumanprimaryendometrialstromalcellsbythewntbcateninsignallingpathway
AT yuanlihe sisnhg5foxf2inhibitstgfb1inducedfibrosisinhumanprimaryendometrialstromalcellsbythewntbcateninsignallingpathway
_version_ 1724422848650412032
spelling doaj-88aba6a5bb38493585cf2b1d19ba9e7a2020-11-25T04:09:12ZengBMCStem Cell Research & Therapy1757-65122020-11-0111111710.1186/s13287-020-01990-3si-SNHG5-FOXF2 inhibits TGF-β1-induced fibrosis in human primary endometrial stromal cells by the Wnt/β-catenin signalling pathwayLimin Liu0Guobin Chen1Taoliang Chen2Wenjuan Shi3Haiyan Hu4Kaijing Song5Ruichun Huang6Huihua Cai7Yuanli He8Department of Obstetrics and Gynecology, Zhujiang Hospital, Southern Medical UniversityDepartment of Obstetrics and Gynecology, Affiliated Shenzhen Maternity & Child Healthcare Hospital, Southern Medical UniversityThe National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Department of Neurosurgery, Zhujiang Hospital, Southern Medical UniversityDepartment of Obstetrics and Gynecology, Affiliated Shenzhen Maternity & Child Healthcare Hospital, Southern Medical UniversityDepartment of Obstetrics and Gynecology, Affiliated Shenzhen Maternity & Child Healthcare Hospital, Southern Medical UniversityDepartment of Obstetrics and Gynecology, Zhujiang Hospital, Southern Medical UniversityDepartment of Obstetrics and Gynecology, Zhujiang Hospital, Southern Medical UniversityDepartment of Obstetrics and Gynecology, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical SciencesDepartment of Obstetrics and Gynecology, Zhujiang Hospital, Southern Medical UniversityAbstract Background Intrauterine adhesions (IUAs) are manifestations of endometrial fibrosis characterized by inflammation and fibrinogen aggregation in the extracellular matrix (ECM). The available therapeutic interventions for IUA are insufficiently effective in the clinical setting for postoperative adhesion recurrence and infertility problems. In this study, we investigated whether si-SNHG5-FOXF2 can serve as a molecular mechanism for the inhibition of IUA fibrosis ex vivo. Methods FOXF2, TGF-β1 and collagen expression levels were measured by microarray sequencing analysis in three normal endometrium groups and six IUA patients. We induced primary human endometrial stromal cells (HESCs) into myofibroblasts (MFs) to develop an IUA cell model with various concentrations of TGF-β1 at various times. Downstream target genes of FOXF2 were screened by chromatin immunoprecipitation combined with whole-genome high-throughput sequencing (ChIP-seq). We investigated ECM formation, cell proliferation and Wnt/β-catenin signalling pathway-related proteins in primary HESCs with FOXF2 downregulation by quantitative reverse transcription-polymerase chain reaction (qRT-PCR), western blotting (WB), immunohistochemistry (IHC), flow cytometry, ethylenediurea (EdU) and CCK8 assays. We identified long noncoding RNAs (lncRNA) SNHG5 as the upstream regulatory gene of FOXF2 through RNA immunoprecipitation (RIP), RNA pulldown and fluorescence in situ hybridization (FISH). Finally, we examined FOXF2 expression, ECM formation, cell proliferation and Wnt/β-catenin signalling pathway-related proteins in primary HESCs upon FOXF2 downregulation. Results FOXF2 was highly expressed in the endometrium of patients with IUA. Treatment of primary HESCs with 10 ng/ml TGF-β1 for 72 h was found to be most effective for developing an IUA cell model. FOXF2 regulated multiple downstream target genes, including collagen, vimentin (VIM) and cyclin D2/DK4, by ChIP-seq and ChIP-PCR. FOXF2 downregulation inhibited TGF-β1-mediated primary HESC fibrosis, including ECM formation, cell proliferation and Wnt/β-catenin signalling pathway-related protein expression. We identified lncRNA SNHG5 as an upstream gene that directly regulates FOXF2 by RIP-seq, qRT-PCR, WB and FISH. SNHG5 downregulation suppressed FOXF2 expression in the IUA cell model, resulting in synergistic repression of the Wnt/β-catenin pathway, thereby altering TGF-β1-mediated ECM aggregation in endometrial stromal cells ex vivo. Conclusions Regulation of the Wnt/β-catenin signalling pathway and ECM formation by si-SNHG5-FOXF2 effectively inhibited the profibrotic effect of TGF-β1 on primary HESCs. This finding can provide a molecular basis for antagonizing TGF-β1-mediated fibrosis in primary HESCs.http://link.springer.com/article/10.1186/s13287-020-01990-3IUATGF-β1FOXF2SNHG5ECMFibrosis