Extracellular Fe2+ and Fe3+ modulate osteocytic viability, expression of SOST, RANKL and FGF23, and fluid flow-induced YAP1 nuclear translocation

Abstract Iron overload negatively affects bone mass and strength. However, the impact of iron excess on osteocytes—important bone cells for mechanotransduction and remodeling—is poorly understood. Herein, we examined the effects of iron exposure on osteocytes during their maturation process. We disc...

Full description

Bibliographic Details
Main Authors: Wasutorn Chankamngoen, Saowalak Krungchanuchat, Jirawan Thongbunchoo, Naraporn Sirinonthanawech, Jarinthorn Teerapornpuntakit, Nattapon Panupinthu, Narattaphol Charoenphandhu
Format: Article
Language:English
Published: Nature Portfolio 2023-12-01
Series:Scientific Reports
Online Access:https://doi.org/10.1038/s41598-023-48436-3
_version_ 1797415268272570368
author Wasutorn Chankamngoen
Saowalak Krungchanuchat
Jirawan Thongbunchoo
Naraporn Sirinonthanawech
Jarinthorn Teerapornpuntakit
Nattapon Panupinthu
Narattaphol Charoenphandhu
author_facet Wasutorn Chankamngoen
Saowalak Krungchanuchat
Jirawan Thongbunchoo
Naraporn Sirinonthanawech
Jarinthorn Teerapornpuntakit
Nattapon Panupinthu
Narattaphol Charoenphandhu
author_sort Wasutorn Chankamngoen
collection DOAJ
description Abstract Iron overload negatively affects bone mass and strength. However, the impact of iron excess on osteocytes—important bone cells for mechanotransduction and remodeling—is poorly understood. Herein, we examined the effects of iron exposure on osteocytes during their maturation process. We discovered that iron overload caused apoptosis of osteocytes in early and late stages of differentiation. Notably, the expression of key proteins for iron entry was downregulated during differentiation, suggesting that mature osteocytes were less susceptible to iron toxicity due to limited iron uptake. Furthermore, iron overload also enriched a subpopulation of mature osteocytes, as indicated by increased expression of Dmp1, a gene encoding protein for bone mineralization. These iron-exposed osteocytes expressed high levels of Sost, Tnfsf11 and Fgf23 transcripts. Consistently, we demonstrated that exogenous FGF23 stimulated the formation and survival of osteoclasts, suggesting its regulatory role in bone resorption. In addition, iron overload downregulated the expression of Cx43, a gene encoding gap junction protein in the dendritic processes, and impaired YAP1 nuclear translocation in response to fluid flow in differentiated osteocytes. It can be concluded that iron overload induces cellular adaptation in differentiating osteocytes, resulting in insensitivity to mechanical stimulation and potential disruption of the balance in bone remodeling.
first_indexed 2024-03-09T05:45:15Z
format Article
id doaj.art-9ec18af3547044098cedae3e0e9d1782
institution Directory Open Access Journal
issn 2045-2322
language English
last_indexed 2024-03-09T05:45:15Z
publishDate 2023-12-01
publisher Nature Portfolio
record_format Article
series Scientific Reports
spelling doaj.art-9ec18af3547044098cedae3e0e9d17822023-12-03T12:21:33ZengNature PortfolioScientific Reports2045-23222023-12-0113111310.1038/s41598-023-48436-3Extracellular Fe2+ and Fe3+ modulate osteocytic viability, expression of SOST, RANKL and FGF23, and fluid flow-induced YAP1 nuclear translocationWasutorn Chankamngoen0Saowalak Krungchanuchat1Jirawan Thongbunchoo2Naraporn Sirinonthanawech3Jarinthorn Teerapornpuntakit4Nattapon Panupinthu5Narattaphol Charoenphandhu6Graduate Program in Molecular Medicine, Faculty of Science, Mahidol UniversityCenter of Calcium and Bone Research (COCAB), Faculty of Science, Mahidol UniversityCenter of Calcium and Bone Research (COCAB), Faculty of Science, Mahidol UniversityInstitute of Molecular Biosciences, Mahidol UniversityCenter of Calcium and Bone Research (COCAB), Faculty of Science, Mahidol UniversityCenter of Calcium and Bone Research (COCAB), Faculty of Science, Mahidol UniversityCenter of Calcium and Bone Research (COCAB), Faculty of Science, Mahidol UniversityAbstract Iron overload negatively affects bone mass and strength. However, the impact of iron excess on osteocytes—important bone cells for mechanotransduction and remodeling—is poorly understood. Herein, we examined the effects of iron exposure on osteocytes during their maturation process. We discovered that iron overload caused apoptosis of osteocytes in early and late stages of differentiation. Notably, the expression of key proteins for iron entry was downregulated during differentiation, suggesting that mature osteocytes were less susceptible to iron toxicity due to limited iron uptake. Furthermore, iron overload also enriched a subpopulation of mature osteocytes, as indicated by increased expression of Dmp1, a gene encoding protein for bone mineralization. These iron-exposed osteocytes expressed high levels of Sost, Tnfsf11 and Fgf23 transcripts. Consistently, we demonstrated that exogenous FGF23 stimulated the formation and survival of osteoclasts, suggesting its regulatory role in bone resorption. In addition, iron overload downregulated the expression of Cx43, a gene encoding gap junction protein in the dendritic processes, and impaired YAP1 nuclear translocation in response to fluid flow in differentiated osteocytes. It can be concluded that iron overload induces cellular adaptation in differentiating osteocytes, resulting in insensitivity to mechanical stimulation and potential disruption of the balance in bone remodeling.https://doi.org/10.1038/s41598-023-48436-3
spellingShingle Wasutorn Chankamngoen
Saowalak Krungchanuchat
Jirawan Thongbunchoo
Naraporn Sirinonthanawech
Jarinthorn Teerapornpuntakit
Nattapon Panupinthu
Narattaphol Charoenphandhu
Extracellular Fe2+ and Fe3+ modulate osteocytic viability, expression of SOST, RANKL and FGF23, and fluid flow-induced YAP1 nuclear translocation
Scientific Reports
title Extracellular Fe2+ and Fe3+ modulate osteocytic viability, expression of SOST, RANKL and FGF23, and fluid flow-induced YAP1 nuclear translocation
title_full Extracellular Fe2+ and Fe3+ modulate osteocytic viability, expression of SOST, RANKL and FGF23, and fluid flow-induced YAP1 nuclear translocation
title_fullStr Extracellular Fe2+ and Fe3+ modulate osteocytic viability, expression of SOST, RANKL and FGF23, and fluid flow-induced YAP1 nuclear translocation
title_full_unstemmed Extracellular Fe2+ and Fe3+ modulate osteocytic viability, expression of SOST, RANKL and FGF23, and fluid flow-induced YAP1 nuclear translocation
title_short Extracellular Fe2+ and Fe3+ modulate osteocytic viability, expression of SOST, RANKL and FGF23, and fluid flow-induced YAP1 nuclear translocation
title_sort extracellular fe2 and fe3 modulate osteocytic viability expression of sost rankl and fgf23 and fluid flow induced yap1 nuclear translocation
url https://doi.org/10.1038/s41598-023-48436-3
work_keys_str_mv AT wasutornchankamngoen extracellularfe2andfe3modulateosteocyticviabilityexpressionofsostranklandfgf23andfluidflowinducedyap1nucleartranslocation
AT saowalakkrungchanuchat extracellularfe2andfe3modulateosteocyticviabilityexpressionofsostranklandfgf23andfluidflowinducedyap1nucleartranslocation
AT jirawanthongbunchoo extracellularfe2andfe3modulateosteocyticviabilityexpressionofsostranklandfgf23andfluidflowinducedyap1nucleartranslocation
AT narapornsirinonthanawech extracellularfe2andfe3modulateosteocyticviabilityexpressionofsostranklandfgf23andfluidflowinducedyap1nucleartranslocation
AT jarinthornteerapornpuntakit extracellularfe2andfe3modulateosteocyticviabilityexpressionofsostranklandfgf23andfluidflowinducedyap1nucleartranslocation
AT nattaponpanupinthu extracellularfe2andfe3modulateosteocyticviabilityexpressionofsostranklandfgf23andfluidflowinducedyap1nucleartranslocation
AT narattapholcharoenphandhu extracellularfe2andfe3modulateosteocyticviabilityexpressionofsostranklandfgf23andfluidflowinducedyap1nucleartranslocation