Fanca deficiency is associated with alterations in osteoclastogenesis that are rescued by TNFα

Abstract Background Hematopoietic stem cells (HSCs) reside in the bone marrow (BM) niche, which includes bone-forming and bone-resorbing cells, i.e., osteoblasts (OBs) and osteoclasts (OCs). OBs originate from mesenchymal progenitors, while OCs are derived from HSCs. Self-renewal, proliferation and...

Full description

Bibliographic Details
Main Authors: Alessia Oppezzo, Lovely Monney, Henri Kilian, Lofti Slimani, Frédérique Maczkowiak-Chartois, Filippo Rosselli
Format: Article
Language:English
Published: BMC 2023-06-01
Series:Cell & Bioscience
Subjects:
Online Access:https://doi.org/10.1186/s13578-023-01067-7
_version_ 1797795473591894016
author Alessia Oppezzo
Lovely Monney
Henri Kilian
Lofti Slimani
Frédérique Maczkowiak-Chartois
Filippo Rosselli
author_facet Alessia Oppezzo
Lovely Monney
Henri Kilian
Lofti Slimani
Frédérique Maczkowiak-Chartois
Filippo Rosselli
author_sort Alessia Oppezzo
collection DOAJ
description Abstract Background Hematopoietic stem cells (HSCs) reside in the bone marrow (BM) niche, which includes bone-forming and bone-resorbing cells, i.e., osteoblasts (OBs) and osteoclasts (OCs). OBs originate from mesenchymal progenitors, while OCs are derived from HSCs. Self-renewal, proliferation and differentiation of HSCs are under the control of regulatory signals generated by OBs and OCs within the BM niche. Consequently, OBs and OCs control both bone physiology and hematopoiesis. Since the human developmental and bone marrow failure genetic syndrome fanconi anemia (FA) presents with skeletal abnormalities, osteoporosis and HSC impairment, we wanted to test the hypothesis that the main pathological abnormalities of FA could be related to a defect in OC physiology and/or in bone homeostasis. Results We revealed here that the intrinsic differentiation of OCs from a Fanca −/− mouse is impaired in vitro due to overactivation of the p53–p21 axis and defects in NF-kB signaling. The OC differentiation abnormalities observed in vitro were rescued by treating Fanca −/− cells with the p53 inhibitor pifithrin-α, by treatment with the proinflammatory cytokine TNFα or by coculturing them with Fanca-proficient or Fanca-deficient osteoblastic cells. Conclusions Overall, our results highlight an unappreciated role of Fanca in OC differentiation that is potentially circumvented in vivo by the presence of OBs and TNFα in the BM niche.
first_indexed 2024-03-13T03:18:30Z
format Article
id doaj.art-bfd377a506b04c929a5c17294a790623
institution Directory Open Access Journal
issn 2045-3701
language English
last_indexed 2024-03-13T03:18:30Z
publishDate 2023-06-01
publisher BMC
record_format Article
series Cell & Bioscience
spelling doaj.art-bfd377a506b04c929a5c17294a7906232023-06-25T11:30:26ZengBMCCell & Bioscience2045-37012023-06-0113111710.1186/s13578-023-01067-7Fanca deficiency is associated with alterations in osteoclastogenesis that are rescued by TNFαAlessia Oppezzo0Lovely Monney1Henri Kilian2Lofti Slimani3Frédérique Maczkowiak-Chartois4Filippo Rosselli5CNRS UMR9019, Équipe labellisée La Ligue contre le CancerCNRS UMR9019, Équipe labellisée La Ligue contre le CancerURP2496 Pathologies, Imagerie et Biothérapies Orofaciales et Plateforme Imagerie du Vivant (PIV), FHU-DDS-net, Dental School, Université de ParisURP2496 Pathologies, Imagerie et Biothérapies Orofaciales et Plateforme Imagerie du Vivant (PIV), FHU-DDS-net, Dental School, Université de ParisCNRS UMR9019, Équipe labellisée La Ligue contre le CancerCNRS UMR9019, Équipe labellisée La Ligue contre le CancerAbstract Background Hematopoietic stem cells (HSCs) reside in the bone marrow (BM) niche, which includes bone-forming and bone-resorbing cells, i.e., osteoblasts (OBs) and osteoclasts (OCs). OBs originate from mesenchymal progenitors, while OCs are derived from HSCs. Self-renewal, proliferation and differentiation of HSCs are under the control of regulatory signals generated by OBs and OCs within the BM niche. Consequently, OBs and OCs control both bone physiology and hematopoiesis. Since the human developmental and bone marrow failure genetic syndrome fanconi anemia (FA) presents with skeletal abnormalities, osteoporosis and HSC impairment, we wanted to test the hypothesis that the main pathological abnormalities of FA could be related to a defect in OC physiology and/or in bone homeostasis. Results We revealed here that the intrinsic differentiation of OCs from a Fanca −/− mouse is impaired in vitro due to overactivation of the p53–p21 axis and defects in NF-kB signaling. The OC differentiation abnormalities observed in vitro were rescued by treating Fanca −/− cells with the p53 inhibitor pifithrin-α, by treatment with the proinflammatory cytokine TNFα or by coculturing them with Fanca-proficient or Fanca-deficient osteoblastic cells. Conclusions Overall, our results highlight an unappreciated role of Fanca in OC differentiation that is potentially circumvented in vivo by the presence of OBs and TNFα in the BM niche.https://doi.org/10.1186/s13578-023-01067-7Fanconi anemiaOsteoclastOsteoblastCell signalingTNFαp53
spellingShingle Alessia Oppezzo
Lovely Monney
Henri Kilian
Lofti Slimani
Frédérique Maczkowiak-Chartois
Filippo Rosselli
Fanca deficiency is associated with alterations in osteoclastogenesis that are rescued by TNFα
Cell & Bioscience
Fanconi anemia
Osteoclast
Osteoblast
Cell signaling
TNFα
p53
title Fanca deficiency is associated with alterations in osteoclastogenesis that are rescued by TNFα
title_full Fanca deficiency is associated with alterations in osteoclastogenesis that are rescued by TNFα
title_fullStr Fanca deficiency is associated with alterations in osteoclastogenesis that are rescued by TNFα
title_full_unstemmed Fanca deficiency is associated with alterations in osteoclastogenesis that are rescued by TNFα
title_short Fanca deficiency is associated with alterations in osteoclastogenesis that are rescued by TNFα
title_sort fanca deficiency is associated with alterations in osteoclastogenesis that are rescued by tnfα
topic Fanconi anemia
Osteoclast
Osteoblast
Cell signaling
TNFα
p53
url https://doi.org/10.1186/s13578-023-01067-7
work_keys_str_mv AT alessiaoppezzo fancadeficiencyisassociatedwithalterationsinosteoclastogenesisthatarerescuedbytnfa
AT lovelymonney fancadeficiencyisassociatedwithalterationsinosteoclastogenesisthatarerescuedbytnfa
AT henrikilian fancadeficiencyisassociatedwithalterationsinosteoclastogenesisthatarerescuedbytnfa
AT loftislimani fancadeficiencyisassociatedwithalterationsinosteoclastogenesisthatarerescuedbytnfa
AT frederiquemaczkowiakchartois fancadeficiencyisassociatedwithalterationsinosteoclastogenesisthatarerescuedbytnfa
AT filipporosselli fancadeficiencyisassociatedwithalterationsinosteoclastogenesisthatarerescuedbytnfa