CD13 is a critical regulator of cell–cell fusion in osteoclastogenesis

Abstract The transmembrane aminopeptidase CD13 is highly expressed in cells of the myeloid lineage, regulates dynamin-dependent receptor endocytosis and recycling and is a necessary component of actin cytoskeletal organization. Here, we show that CD13-deficient mice present a low bone density phenot...

Full description

Bibliographic Details
Main Authors: Mallika Ghosh, Tomislav Kelava, Ivana Vrhovac Madunic, Ivo Kalajzic, Linda H. Shapiro
Format: Article
Language:English
Published: Nature Publishing Group 2021-05-01
Series:Scientific Reports
Online Access:https://doi.org/10.1038/s41598-021-90271-x
id doaj-c76605703dea4829b0ced176c41a2067
record_format Article
spelling doaj-c76605703dea4829b0ced176c41a20672021-05-30T11:38:19ZengNature Publishing GroupScientific Reports2045-23222021-05-0111111410.1038/s41598-021-90271-xCD13 is a critical regulator of cell–cell fusion in osteoclastogenesisMallika Ghosh0Tomislav Kelava1Ivana Vrhovac Madunic2Ivo Kalajzic3Linda H. Shapiro4Center for Vascular Biology, University of Connecticut Medical SchoolCenter for Regenerative Medicine and Skeletal Development, University of Connecticut Dental SchoolCenter for Regenerative Medicine and Skeletal Development, University of Connecticut Dental SchoolCenter for Regenerative Medicine and Skeletal Development, University of Connecticut Dental SchoolCenter for Vascular Biology, University of Connecticut Medical SchoolAbstract The transmembrane aminopeptidase CD13 is highly expressed in cells of the myeloid lineage, regulates dynamin-dependent receptor endocytosis and recycling and is a necessary component of actin cytoskeletal organization. Here, we show that CD13-deficient mice present a low bone density phenotype with increased numbers of osteoclasts per bone surface, but display a normal distribution of osteoclast progenitor populations in the bone marrow and periphery. In addition, the bone formation and mineral apposition rates are similar between genotypes, indicating a defect in osteoclast-specific function in vivo. Lack of CD13 led to exaggerated in vitro osteoclastogenesis as indicated by significantly enhanced fusion of bone marrow-derived multinucleated osteoclasts in the presence of M-CSF and RANKL, resulting in abnormally large cells containing remarkably high numbers of nuclei. Mechanistically, while expression levels of the fusion-regulatory proteins dynamin and DC-STAMP1 must be downregulated for fusion to proceed, these are aberrantly sustained at high levels even in CD13-deficient mature multi-nucleated osteoclasts. Further, the stability of fusion-promoting proteins is maintained in the absence of CD13, implicating CD13 in protein turnover mechanisms. Together, we conclude that CD13 may regulate cell–cell fusion by controlling the expression and localization of key fusion regulatory proteins that are critical for osteoclast fusion.https://doi.org/10.1038/s41598-021-90271-x
collection DOAJ
language English
format Article
sources DOAJ
author Mallika Ghosh
Tomislav Kelava
Ivana Vrhovac Madunic
Ivo Kalajzic
Linda H. Shapiro
spellingShingle Mallika Ghosh
Tomislav Kelava
Ivana Vrhovac Madunic
Ivo Kalajzic
Linda H. Shapiro
CD13 is a critical regulator of cell–cell fusion in osteoclastogenesis
Scientific Reports
author_facet Mallika Ghosh
Tomislav Kelava
Ivana Vrhovac Madunic
Ivo Kalajzic
Linda H. Shapiro
author_sort Mallika Ghosh
title CD13 is a critical regulator of cell–cell fusion in osteoclastogenesis
title_short CD13 is a critical regulator of cell–cell fusion in osteoclastogenesis
title_full CD13 is a critical regulator of cell–cell fusion in osteoclastogenesis
title_fullStr CD13 is a critical regulator of cell–cell fusion in osteoclastogenesis
title_full_unstemmed CD13 is a critical regulator of cell–cell fusion in osteoclastogenesis
title_sort cd13 is a critical regulator of cell–cell fusion in osteoclastogenesis
publisher Nature Publishing Group
series Scientific Reports
issn 2045-2322
publishDate 2021-05-01
description Abstract The transmembrane aminopeptidase CD13 is highly expressed in cells of the myeloid lineage, regulates dynamin-dependent receptor endocytosis and recycling and is a necessary component of actin cytoskeletal organization. Here, we show that CD13-deficient mice present a low bone density phenotype with increased numbers of osteoclasts per bone surface, but display a normal distribution of osteoclast progenitor populations in the bone marrow and periphery. In addition, the bone formation and mineral apposition rates are similar between genotypes, indicating a defect in osteoclast-specific function in vivo. Lack of CD13 led to exaggerated in vitro osteoclastogenesis as indicated by significantly enhanced fusion of bone marrow-derived multinucleated osteoclasts in the presence of M-CSF and RANKL, resulting in abnormally large cells containing remarkably high numbers of nuclei. Mechanistically, while expression levels of the fusion-regulatory proteins dynamin and DC-STAMP1 must be downregulated for fusion to proceed, these are aberrantly sustained at high levels even in CD13-deficient mature multi-nucleated osteoclasts. Further, the stability of fusion-promoting proteins is maintained in the absence of CD13, implicating CD13 in protein turnover mechanisms. Together, we conclude that CD13 may regulate cell–cell fusion by controlling the expression and localization of key fusion regulatory proteins that are critical for osteoclast fusion.
url https://doi.org/10.1038/s41598-021-90271-x
work_keys_str_mv AT mallikaghosh cd13isacriticalregulatorofcellcellfusioninosteoclastogenesis
AT tomislavkelava cd13isacriticalregulatorofcellcellfusioninosteoclastogenesis
AT ivanavrhovacmadunic cd13isacriticalregulatorofcellcellfusioninosteoclastogenesis
AT ivokalajzic cd13isacriticalregulatorofcellcellfusioninosteoclastogenesis
AT lindahshapiro cd13isacriticalregulatorofcellcellfusioninosteoclastogenesis
_version_ 1721420192583516160