Your browser doesn't support javascript.
loading
ATP6AP2, a regulator of LRP6/ß-catenin protein trafficking, promotes Wnt/ß-catenin signaling and bone formation in a cell type dependent manner.
Xiong, Lei; Guo, Hao-Han; Pan, Jin-Xiu; Ren, Xiao; Lee, Daehoon; Chen, Li; Mei, Lin; Xiong, Wen-Cheng.
Afiliación
  • Xiong L; Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, OH, 44106, USA.
  • Guo HH; Louis Stoke VA Medical Center, Cleveland, OH, 44106, USA.
  • Pan JX; Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, OH, 44106, USA.
  • Ren X; Louis Stoke VA Medical Center, Cleveland, OH, 44106, USA.
  • Lee D; Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, OH, 44106, USA.
  • Chen L; Louis Stoke VA Medical Center, Cleveland, OH, 44106, USA.
  • Mei L; Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, OH, 44106, USA.
  • Xiong WC; Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, OH, 44106, USA.
Bone Res ; 12(1): 33, 2024 May 29.
Article en En | MEDLINE | ID: mdl-38811544
ABSTRACT
Wnt/ß-catenin signaling is critical for various cellular processes in multiple cell types, including osteoblast (OB) differentiation and function. Exactly how Wnt/ß-catenin signaling is regulated in OBs remain elusive. ATP6AP2, an accessory subunit of V-ATPase, plays important roles in multiple cell types/organs and multiple signaling pathways. However, little is known whether and how ATP6AP2 in OBs regulates Wnt/ß-catenin signaling and bone formation. Here we provide evidence for ATP6AP2 in the OB-lineage cells to promote OB-mediated bone formation and bone homeostasis selectively in the trabecular bone regions. Conditionally knocking out (CKO) ATP6AP2 in the OB-lineage cells (Atp6ap2Ocn-Cre) reduced trabecular, but not cortical, bone formation and bone mass. Proteomic and cellular biochemical studies revealed that LRP6 and N-cadherin were reduced in ATP6AP2-KO BMSCs and OBs, but not osteocytes. Additional in vitro and in vivo studies revealed impaired ß-catenin signaling in ATP6AP2-KO BMSCs and OBs, but not osteocytes, under both basal and Wnt stimulated conditions, although LRP5 was decreased in ATP6AP2-KO osteocytes, but not BMSCs. Further cell biological studies uncovered that osteoblastic ATP6AP2 is not required for Wnt3a suppression of ß-catenin phosphorylation, but necessary for LRP6/ß-catenin and N-cadherin/ß-catenin protein complex distribution at the cell membrane, thus preventing their degradation. Expression of active ß-catenin diminished the OB differentiation deficit in ATP6AP2-KO BMSCs. Taken together, these results support the view for ATP6AP2 as a critical regulator of both LRP6 and N-cadherin protein trafficking and stability, and thus regulating ß-catenin levels, demonstrating an un-recognized function of osteoblastic ATP6AP2 in promoting Wnt/LRP6/ß-catenin signaling and trabecular bone formation.
Asunto(s)

Texto completo: 1 Banco de datos: MEDLINE Asunto principal: Osteoblastos / Osteogénesis / Ratones Noqueados / ATPasas de Translocación de Protón Vacuolares / Beta Catenina / Proteína-6 Relacionada a Receptor de Lipoproteína de Baja Densidad / Vía de Señalización Wnt Límite: Animals Idioma: En Revista: Bone Res Año: 2024 Tipo del documento: Article País de afiliación: Estados Unidos

Texto completo: 1 Banco de datos: MEDLINE Asunto principal: Osteoblastos / Osteogénesis / Ratones Noqueados / ATPasas de Translocación de Protón Vacuolares / Beta Catenina / Proteína-6 Relacionada a Receptor de Lipoproteína de Baja Densidad / Vía de Señalización Wnt Límite: Animals Idioma: En Revista: Bone Res Año: 2024 Tipo del documento: Article País de afiliación: Estados Unidos