Cargando…

Effects of fluoride on the proliferation and activation of osteoblasts by regulating methylation of the DNA repair genes MGMT and MLH1

INTRODUCTION: Fluoride can induce the proliferation and activation of osteoblasts, resulting in skeletal fluorosis progression; however, the specific mechanism is unclear. METHODS: Cell proliferation was examined using the MTT assay. Flow cytometry was performed to detect the cell cycle distribution...

Descripción completa

Detalles Bibliográficos
Autores principales: Chen, Long, Yin, Na, Ding, Yi, Zhang, Mei-Lin, Li, Min, Zhong, Jin-Jie, Feng, Shu-Mei
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Japanese Society for Regenerative Medicine 2022
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8814769/
https://www.ncbi.nlm.nih.gov/pubmed/35155723
http://dx.doi.org/10.1016/j.reth.2022.01.004
_version_ 1784645135374483456
author Chen, Long
Yin, Na
Ding, Yi
Zhang, Mei-Lin
Li, Min
Zhong, Jin-Jie
Feng, Shu-Mei
author_facet Chen, Long
Yin, Na
Ding, Yi
Zhang, Mei-Lin
Li, Min
Zhong, Jin-Jie
Feng, Shu-Mei
author_sort Chen, Long
collection PubMed
description INTRODUCTION: Fluoride can induce the proliferation and activation of osteoblasts, resulting in skeletal fluorosis progression; however, the specific mechanism is unclear. METHODS: Cell proliferation was examined using the MTT assay. Flow cytometry was performed to detect the cell cycle distribution. Alkaline phosphatase (ALP) was calculated to evaluate bone formation and turnover. Gene methylation was examined using the MSP assay. mRNA and protein expression levels were assessed using qRT-PCR and Western blot assays. RESULTS: Low-concentration NaF treatment promoted the cell cycle progression of osteoblasts to S-phase, thus accelerating cell proliferation and activation in a concentration-dependent manner. In addition, the methylation of the MGMT and MLH1 genes was increased, and their mRNA expression was reduced. Furthermore, the DNA methyltransferase inhibitor 5-AZA-dC suppressed cell viability, cell number in S-phase, ALP activity and osteogenesis-related protein levels in osteoblasts treated with low doses of NaF. Meanwhile, 5-AZA-dC suppressed the increase in MGMT and MLH1 gene methylation in osteoblasts treated with low doses of NaF, leading to enhanced expression of MGMT and MLH1 mRNA. CONCLUSION: NaF treatment led to methylation of the DNA repair genes MGMT and MLH1 in osteoblasts, resulting in cell proliferation and activation and causing the development of skeletal fluorosis.
format Online
Article
Text
id pubmed-8814769
institution National Center for Biotechnology Information
language English
publishDate 2022
publisher Japanese Society for Regenerative Medicine
record_format MEDLINE/PubMed
spelling pubmed-88147692022-02-11 Effects of fluoride on the proliferation and activation of osteoblasts by regulating methylation of the DNA repair genes MGMT and MLH1 Chen, Long Yin, Na Ding, Yi Zhang, Mei-Lin Li, Min Zhong, Jin-Jie Feng, Shu-Mei Regen Ther Original Article INTRODUCTION: Fluoride can induce the proliferation and activation of osteoblasts, resulting in skeletal fluorosis progression; however, the specific mechanism is unclear. METHODS: Cell proliferation was examined using the MTT assay. Flow cytometry was performed to detect the cell cycle distribution. Alkaline phosphatase (ALP) was calculated to evaluate bone formation and turnover. Gene methylation was examined using the MSP assay. mRNA and protein expression levels were assessed using qRT-PCR and Western blot assays. RESULTS: Low-concentration NaF treatment promoted the cell cycle progression of osteoblasts to S-phase, thus accelerating cell proliferation and activation in a concentration-dependent manner. In addition, the methylation of the MGMT and MLH1 genes was increased, and their mRNA expression was reduced. Furthermore, the DNA methyltransferase inhibitor 5-AZA-dC suppressed cell viability, cell number in S-phase, ALP activity and osteogenesis-related protein levels in osteoblasts treated with low doses of NaF. Meanwhile, 5-AZA-dC suppressed the increase in MGMT and MLH1 gene methylation in osteoblasts treated with low doses of NaF, leading to enhanced expression of MGMT and MLH1 mRNA. CONCLUSION: NaF treatment led to methylation of the DNA repair genes MGMT and MLH1 in osteoblasts, resulting in cell proliferation and activation and causing the development of skeletal fluorosis. Japanese Society for Regenerative Medicine 2022-01-30 /pmc/articles/PMC8814769/ /pubmed/35155723 http://dx.doi.org/10.1016/j.reth.2022.01.004 Text en © 2022 The Japanese Society for Regenerative Medicine. Production and hosting by Elsevier B.V. https://creativecommons.org/licenses/by-nc-nd/4.0/This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/).
spellingShingle Original Article
Chen, Long
Yin, Na
Ding, Yi
Zhang, Mei-Lin
Li, Min
Zhong, Jin-Jie
Feng, Shu-Mei
Effects of fluoride on the proliferation and activation of osteoblasts by regulating methylation of the DNA repair genes MGMT and MLH1
title Effects of fluoride on the proliferation and activation of osteoblasts by regulating methylation of the DNA repair genes MGMT and MLH1
title_full Effects of fluoride on the proliferation and activation of osteoblasts by regulating methylation of the DNA repair genes MGMT and MLH1
title_fullStr Effects of fluoride on the proliferation and activation of osteoblasts by regulating methylation of the DNA repair genes MGMT and MLH1
title_full_unstemmed Effects of fluoride on the proliferation and activation of osteoblasts by regulating methylation of the DNA repair genes MGMT and MLH1
title_short Effects of fluoride on the proliferation and activation of osteoblasts by regulating methylation of the DNA repair genes MGMT and MLH1
title_sort effects of fluoride on the proliferation and activation of osteoblasts by regulating methylation of the dna repair genes mgmt and mlh1
topic Original Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8814769/
https://www.ncbi.nlm.nih.gov/pubmed/35155723
http://dx.doi.org/10.1016/j.reth.2022.01.004
work_keys_str_mv AT chenlong effectsoffluorideontheproliferationandactivationofosteoblastsbyregulatingmethylationofthednarepairgenesmgmtandmlh1
AT yinna effectsoffluorideontheproliferationandactivationofosteoblastsbyregulatingmethylationofthednarepairgenesmgmtandmlh1
AT dingyi effectsoffluorideontheproliferationandactivationofosteoblastsbyregulatingmethylationofthednarepairgenesmgmtandmlh1
AT zhangmeilin effectsoffluorideontheproliferationandactivationofosteoblastsbyregulatingmethylationofthednarepairgenesmgmtandmlh1
AT limin effectsoffluorideontheproliferationandactivationofosteoblastsbyregulatingmethylationofthednarepairgenesmgmtandmlh1
AT zhongjinjie effectsoffluorideontheproliferationandactivationofosteoblastsbyregulatingmethylationofthednarepairgenesmgmtandmlh1
AT fengshumei effectsoffluorideontheproliferationandactivationofosteoblastsbyregulatingmethylationofthednarepairgenesmgmtandmlh1