Osteocytes as main responders to low-intensity pulsed ultrasound treatment during fracture healing

Author: Tatsuya Shimizu1,2, Naomasa Fujita1,3, Kiyomi Tsuji-Tamura1, Yoshimasa Kitagawa2, Toshiaki Fujisawa3, Masato Tamura1, Mari Sato4
Affiliation:
1 Oral Biochemistry and Molecular Biology, Graduate School of Dental Medicine, Hokkaido University, Kita 13, Nishi 7, Kita-ku, Sapporo, 060-8586, Japan.
2 Oral Diagnosis and Medicine, Graduate School of Dental Medicine, Hokkaido University, Sapporo, Japan.
3 Dental Anesthesiology, Graduate School of Dental Medicine, Hokkaido University, Sapporo, Japan.
4 Oral Biochemistry and Molecular Biology, Graduate School of Dental Medicine, Hokkaido University, Kita 13, Nishi 7, Kita-ku, Sapporo, 060-8586, Japan. satomari@den.hokudai.ac.jp.
Conference/Journal: Sci Rep
Date published: 2021 May 13
Other: Volume ID: 11 , Issue ID: 1 , Pages: 10298 , Special Notes: doi: 10.1038/s41598-021-89672-9. , Word Count: 223


Ultrasound stimulation is a type of mechanical stress, and low-intensity pulsed ultrasound (LIPUS) devices have been used clinically to promote fracture healing. However, it remains unclear which skeletal cells, in particular osteocytes or osteoblasts, primarily respond to LIPUS stimulation and how they contribute to fracture healing. To examine this, we utilized medaka, whose bone lacks osteocytes, and zebrafish, whose bone has osteocytes, as in vivo models. Fracture healing was accelerated by ultrasound stimulation in zebrafish, but not in medaka. To examine the molecular events induced by LIPUS stimulation in osteocytes, we performed RNA sequencing of a murine osteocytic cell line exposed to LIPUS. 179 genes reacted to LIPUS stimulation, and functional cluster analysis identified among them several molecular signatures related to immunity, secretion, and transcription. Notably, most of the isolated transcription-related genes were also modulated by LIPUS in vivo in zebrafish. However, expression levels of early growth response protein 1 and 2 (Egr1, 2), JunB, forkhead box Q1 (FoxQ1), and nuclear factor of activated T cells c1 (NFATc1) were not altered by LIPUS in medaka, suggesting that these genes are key transcriptional regulators of LIPUS-dependent fracture healing via osteocytes. We therefore show that bone-embedded osteocytes are necessary for LIPUS-induced promotion of fracture healing via transcriptional control of target genes, which presumably activates neighboring cells involved in fracture healing processes.


PMID: 33986415 PMCID: PMC8119462 DOI: 10.1038/s41598-021-89672-9

BACK