Cargando…

LncRNA‐AK137033 inhibits the osteogenic potential of adipose‐derived stem cells in diabetic osteoporosis by regulating Wnt signaling pathway via DNA methylation

OBJECTIVES: Bone tissue engineering based on adipose‐derived stem cells (ASCs) is expected to become a new treatment for diabetic osteoporosis (DOP) patients with bone defects. However, compared with control ASCs (CON‐ASCs), osteogenic potential of DOP‐ASCs is decreased, which increased the difficul...

Descripción completa

Detalles Bibliográficos
Autores principales: Peng, Shuanglin, Gao, Yujin, Shi, Sirong, Zhao, Dan, Cao, Huayue, Fu, Ting, Cai, Xiaoxiao, Xiao, Jingang
Formato: Online Artículo Texto
Lenguaje:English
Publicado: John Wiley and Sons Inc. 2021
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8780896/
https://www.ncbi.nlm.nih.gov/pubmed/34953002
http://dx.doi.org/10.1111/cpr.13174
_version_ 1784637955733716992
author Peng, Shuanglin
Gao, Yujin
Shi, Sirong
Zhao, Dan
Cao, Huayue
Fu, Ting
Cai, Xiaoxiao
Xiao, Jingang
author_facet Peng, Shuanglin
Gao, Yujin
Shi, Sirong
Zhao, Dan
Cao, Huayue
Fu, Ting
Cai, Xiaoxiao
Xiao, Jingang
author_sort Peng, Shuanglin
collection PubMed
description OBJECTIVES: Bone tissue engineering based on adipose‐derived stem cells (ASCs) is expected to become a new treatment for diabetic osteoporosis (DOP) patients with bone defects. However, compared with control ASCs (CON‐ASCs), osteogenic potential of DOP‐ASCs is decreased, which increased the difficulty of bone reconstruction in DOP patients. Moreover, the cause of the poor osteogenesis of ASCs in a hyperglycemic microenvironment has not been elucidated. Therefore, this study explored the molecular mechanism of the decline in the osteogenic potential of DOP‐ASCs from the perspective of epigenetics to provide a possible therapeutic target for bone repair in DOP patients with bone defects. MATERIALS AND METHODS: An animal model of DOP was established in mice. CON‐ASCs and DOP‐ASCs were isolated from CON and DOP mice, respectively. AK137033 small interfering RNA (SiRNA) and an AK137033 overexpression plasmid were used to regulate the expression of AK137033 in CON‐ASCs and DOP‐ASCs in vitro. Lentiviruses that carried shRNA‐AK137033 or AK137033 cDNA were used to knockdown or overexpress AK137033, respectively, in CON‐ASCs and DOP‐ASCs in vivo. Hematoxylin and eosin (H&E), Masson's, alizarin red, and alkaline phosphatase (ALP) staining, micro‐computed tomography (Micro‐CT), flow cytometry, qPCR, western blotting, immunofluorescence, and bisulfite‐specific PCR (BSP) were used to analyze the functional changes of ASCs. RESULTS: The DOP mouse model was established successfully. Compared with CON‐ASCs, AK137033 expression, the DNA methylation level of the sFrp2 promoter region, Wnt signaling pathway markers, and the osteogenic differentiation potential were decreased in DOP‐ASCs. In vitro experiments showed that AK137033 silencing inhibited the Wnt signaling pathway and osteogenic ability of CON‐ASCs by reducing the DNA methylation level in the sFrp2 promoter region. Additionally, overexpression of AK137033 in DOP‐ASCs rescued these changes caused by DOP. Moreover, the same results were obtained in vivo. CONCLUSIONS: LncRNA‐AK137033 inhibits the osteogenic potential of DOP‐ASCs by regulating the Wnt signaling pathway via modulating the DNA methylation level in the sFrp2 promoter region. This study provides an important reference to find new targets for the treatment of bone defects in DOP patients.
format Online
Article
Text
id pubmed-8780896
institution National Center for Biotechnology Information
language English
publishDate 2021
publisher John Wiley and Sons Inc.
record_format MEDLINE/PubMed
spelling pubmed-87808962022-02-01 LncRNA‐AK137033 inhibits the osteogenic potential of adipose‐derived stem cells in diabetic osteoporosis by regulating Wnt signaling pathway via DNA methylation Peng, Shuanglin Gao, Yujin Shi, Sirong Zhao, Dan Cao, Huayue Fu, Ting Cai, Xiaoxiao Xiao, Jingang Cell Prolif Original Articles OBJECTIVES: Bone tissue engineering based on adipose‐derived stem cells (ASCs) is expected to become a new treatment for diabetic osteoporosis (DOP) patients with bone defects. However, compared with control ASCs (CON‐ASCs), osteogenic potential of DOP‐ASCs is decreased, which increased the difficulty of bone reconstruction in DOP patients. Moreover, the cause of the poor osteogenesis of ASCs in a hyperglycemic microenvironment has not been elucidated. Therefore, this study explored the molecular mechanism of the decline in the osteogenic potential of DOP‐ASCs from the perspective of epigenetics to provide a possible therapeutic target for bone repair in DOP patients with bone defects. MATERIALS AND METHODS: An animal model of DOP was established in mice. CON‐ASCs and DOP‐ASCs were isolated from CON and DOP mice, respectively. AK137033 small interfering RNA (SiRNA) and an AK137033 overexpression plasmid were used to regulate the expression of AK137033 in CON‐ASCs and DOP‐ASCs in vitro. Lentiviruses that carried shRNA‐AK137033 or AK137033 cDNA were used to knockdown or overexpress AK137033, respectively, in CON‐ASCs and DOP‐ASCs in vivo. Hematoxylin and eosin (H&E), Masson's, alizarin red, and alkaline phosphatase (ALP) staining, micro‐computed tomography (Micro‐CT), flow cytometry, qPCR, western blotting, immunofluorescence, and bisulfite‐specific PCR (BSP) were used to analyze the functional changes of ASCs. RESULTS: The DOP mouse model was established successfully. Compared with CON‐ASCs, AK137033 expression, the DNA methylation level of the sFrp2 promoter region, Wnt signaling pathway markers, and the osteogenic differentiation potential were decreased in DOP‐ASCs. In vitro experiments showed that AK137033 silencing inhibited the Wnt signaling pathway and osteogenic ability of CON‐ASCs by reducing the DNA methylation level in the sFrp2 promoter region. Additionally, overexpression of AK137033 in DOP‐ASCs rescued these changes caused by DOP. Moreover, the same results were obtained in vivo. CONCLUSIONS: LncRNA‐AK137033 inhibits the osteogenic potential of DOP‐ASCs by regulating the Wnt signaling pathway via modulating the DNA methylation level in the sFrp2 promoter region. This study provides an important reference to find new targets for the treatment of bone defects in DOP patients. John Wiley and Sons Inc. 2021-12-24 /pmc/articles/PMC8780896/ /pubmed/34953002 http://dx.doi.org/10.1111/cpr.13174 Text en © 2021 The Authors. Cell Proliferation Published by John Wiley & Sons Ltd. https://creativecommons.org/licenses/by/4.0/This is an open access article under the terms of the http://creativecommons.org/licenses/by/4.0/ (https://creativecommons.org/licenses/by/4.0/) License, which permits use, distribution and reproduction in any medium, provided the original work is properly cited.
spellingShingle Original Articles
Peng, Shuanglin
Gao, Yujin
Shi, Sirong
Zhao, Dan
Cao, Huayue
Fu, Ting
Cai, Xiaoxiao
Xiao, Jingang
LncRNA‐AK137033 inhibits the osteogenic potential of adipose‐derived stem cells in diabetic osteoporosis by regulating Wnt signaling pathway via DNA methylation
title LncRNA‐AK137033 inhibits the osteogenic potential of adipose‐derived stem cells in diabetic osteoporosis by regulating Wnt signaling pathway via DNA methylation
title_full LncRNA‐AK137033 inhibits the osteogenic potential of adipose‐derived stem cells in diabetic osteoporosis by regulating Wnt signaling pathway via DNA methylation
title_fullStr LncRNA‐AK137033 inhibits the osteogenic potential of adipose‐derived stem cells in diabetic osteoporosis by regulating Wnt signaling pathway via DNA methylation
title_full_unstemmed LncRNA‐AK137033 inhibits the osteogenic potential of adipose‐derived stem cells in diabetic osteoporosis by regulating Wnt signaling pathway via DNA methylation
title_short LncRNA‐AK137033 inhibits the osteogenic potential of adipose‐derived stem cells in diabetic osteoporosis by regulating Wnt signaling pathway via DNA methylation
title_sort lncrna‐ak137033 inhibits the osteogenic potential of adipose‐derived stem cells in diabetic osteoporosis by regulating wnt signaling pathway via dna methylation
topic Original Articles
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8780896/
https://www.ncbi.nlm.nih.gov/pubmed/34953002
http://dx.doi.org/10.1111/cpr.13174
work_keys_str_mv AT pengshuanglin lncrnaak137033inhibitstheosteogenicpotentialofadiposederivedstemcellsindiabeticosteoporosisbyregulatingwntsignalingpathwayviadnamethylation
AT gaoyujin lncrnaak137033inhibitstheosteogenicpotentialofadiposederivedstemcellsindiabeticosteoporosisbyregulatingwntsignalingpathwayviadnamethylation
AT shisirong lncrnaak137033inhibitstheosteogenicpotentialofadiposederivedstemcellsindiabeticosteoporosisbyregulatingwntsignalingpathwayviadnamethylation
AT zhaodan lncrnaak137033inhibitstheosteogenicpotentialofadiposederivedstemcellsindiabeticosteoporosisbyregulatingwntsignalingpathwayviadnamethylation
AT caohuayue lncrnaak137033inhibitstheosteogenicpotentialofadiposederivedstemcellsindiabeticosteoporosisbyregulatingwntsignalingpathwayviadnamethylation
AT futing lncrnaak137033inhibitstheosteogenicpotentialofadiposederivedstemcellsindiabeticosteoporosisbyregulatingwntsignalingpathwayviadnamethylation
AT caixiaoxiao lncrnaak137033inhibitstheosteogenicpotentialofadiposederivedstemcellsindiabeticosteoporosisbyregulatingwntsignalingpathwayviadnamethylation
AT xiaojingang lncrnaak137033inhibitstheosteogenicpotentialofadiposederivedstemcellsindiabeticosteoporosisbyregulatingwntsignalingpathwayviadnamethylation