Abstract

The loss-of-function mutations in the ALPL result in hypophosphatasia (HPP), an inborn metabolic disorder that causes skeletal mineralization defects. In adults, the main clinical features are early loss of primary or secondary teeth, osteoporosis, bone pain, chondrocalcinosis, and fractures. However, guidelines for the treatment of adults with HPP are not available. Here, we show that ALPL deficiency caused a reduction in intracellular Ca2+ influx, resulting in an osteoporotic phenotype due to downregulated osteogenic differentiation and upregulated adipogenic differentiation in both human and mouse bone marrow mesenchymal stem cells (BMSCs). Increasing the intracellular level of calcium in BMSCs by ionomycin treatment rescued the osteoporotic phenotype in alpl+/− mice and BMSC-specific (Prrx1-alpl−/−) conditional alpl knockout mice. Mechanistically, ALPL was found to be required for the maintenance of intracellular Ca2+ influx, which it achieves by regulating L-type Ca2+ channel trafficking via binding to the α2δ subunits to regulate the internalization of the L-type Ca2+ channel. Decreased Ca2+ flux inactivates the Akt/GSK3β/β-catenin signaling pathway, which regulates lineage differentiation of BMSCs. This study identifies a previously unknown role of the ectoenzyme ALPL in the maintenance of calcium channel trafficking to regulate stem cell lineage differentiation and bone homeostasis. Accelerating Ca2+ flux through L-type Ca2+ channels by ionomycin treatment may be a promising therapeutic approach for adult patients with HPP.

Highlights

  • A loss-of-function mutation in the liver/bone/kidney alkaline phosphatase (ALPL) gene results in the life-threatening disease hypophosphatasia (HPP) during early developmental periods; HPP is characterized by hypomineralization of the skeleton and teeth.[1,2] Adult patients with HPP showed early loss of primary or secondary teeth, osteoporosis, bone pain, chondrocalcinosis, and fractures

  • We found that CaV1.2 and CaV1.3 overlapped with ALPL in Bone marrow mesenchymal stem cells (BMSCs) (Fig. 4a, upper panel), suggesting the association of ALPL and L-type Ca2+ channels

  • We found that the expression of active β-catenin was decreased in both alpl+/− BMSCs and BMSCs transfected with shALP (Fig. 5a)

Read more

Summary

1234567890();,: INTRODUCTION

A loss-of-function mutation in the liver/bone/kidney alkaline phosphatase (ALPL) gene results in the life-threatening disease hypophosphatasia (HPP) during early developmental periods; HPP is characterized by hypomineralization of the skeleton and teeth.[1,2] Adult patients with HPP showed early loss of primary or secondary teeth, osteoporosis, bone pain, chondrocalcinosis, and fractures. The results showed that the total protein expression of CaV1.2 and CaV1.3 was decreased significantly in alpl+/− BMSCs compared with WT BMSCs (Fig. 1d). We membrane protein extraction kit (Abcam, ab65400) to isolate used human and mouse models to demonstrate that ALPL is required for the maintenance of intracellular Ca2+ influx because it regulates L-type Ca2+ channel trafficking via binding to the α2δ subunits, which regulate the internalization of L-type Ca2+ channels Membrane expression of calcium channels affects calcium influx, and we compared the raising the intracellular level of calcium in BMSCs by treatment with ionomycin rescues the osteoporotic phenotype in alpl+/− mice and BMSC-specific (Prrx1-alpl−/−) conditional alpl knockout mice, and the treatment restores stem cell function of BMSCs from HPP patients, suggesting a new strategy for HPP therapy

RESULTS
DISCUSSION
Leti-alp
Findings
MATERIALS AND METHODS
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call