1
|
Yamashita Y, Hayashi M, Liu A, Sasaki F, Tsuchiya Y, Takayanagi H, Saito M, Nakashima T. Fam102a translocates Runx2 and Rbpjl to facilitate Osterix expression and bone formation. Nat Commun 2025; 16:9. [PMID: 39747056 PMCID: PMC11695619 DOI: 10.1038/s41467-024-55451-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Accepted: 12/11/2024] [Indexed: 01/04/2025] Open
Abstract
Bone remodeling maintains the robustness of the bone tissue by balancing bone resorption by osteoclasts and bone formation by osteoblasts. Although these cells together play a crucial role in bone remodeling, only a few reports are available on the common factors involved in the differentiation of the two types of cells. Here, we show family with sequence similarity 102 member A (Fam102a) as a bone-remodeling factor that positively regulates both osteoclast and osteoblast differentiation. Fam102a regulates osteoblast differentiation by controlling recombination signal binding protein for immunoglobulin κ J region-like (Rbpjl). The Fam102a-Rbpjl axis promotes the nuclear translocation of transcription factors and enhances the expression of Osterix, a transcription factor essential for osteoblast differentiation. The deletion of Fam102a or a functional mutation in Rbpjl leads to osteopenia accompanied by reduced osteoblastic bone formation. Thus, the Fam102a-Rbpjl axis plays an important role in osteoblasts and this finding provides insights into bone remodeling.
Collapse
Affiliation(s)
- Yu Yamashita
- Department of Cell Signaling, Graduate School of Medical and Dental Sciences, Institute of Science Tokyo, Tokyo, Japan
- Department of Orthopaedic Surgery, The Jikei University School of Medicine, Tokyo, Japan
| | - Mikihito Hayashi
- Department of Cell Signaling, Graduate School of Medical and Dental Sciences, Institute of Science Tokyo, Tokyo, Japan.
| | - Anhao Liu
- Department of Cell Signaling, Graduate School of Medical and Dental Sciences, Institute of Science Tokyo, Tokyo, Japan
| | - Fumiyuki Sasaki
- Department of Cell Signaling, Graduate School of Medical and Dental Sciences, Institute of Science Tokyo, Tokyo, Japan
| | - Yosuke Tsuchiya
- Department of Cell Signaling, Graduate School of Medical and Dental Sciences, Institute of Science Tokyo, Tokyo, Japan
| | - Hiroshi Takayanagi
- Department of Immunology, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, Tokyo, Japan
| | - Mitsuru Saito
- Department of Orthopaedic Surgery, The Jikei University School of Medicine, Tokyo, Japan
| | | |
Collapse
|
2
|
Zhang J, Liu C, Li J, Yu T, Ruan J, Yang F. Advanced Piezoelectric Materials, Devices, and Systems for Orthopedic Medicine. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2410400. [PMID: 39665130 PMCID: PMC11744659 DOI: 10.1002/advs.202410400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 11/14/2024] [Indexed: 12/13/2024]
Abstract
Harnessing the robust electromechanical couplings, piezoelectric materials not only enable efficient bio-energy harvesting, physiological sensing and actuating but also open enormous opportunities for therapeutic treatments through surface polarization directly interacting with electroactive cells, tissues, and organs. Known for its highly oriented and hierarchical structure, collagen in natural bones produces local electrical signals to stimulate osteoblasts and promote bone formation, inspiring the application of piezoelectric materials in orthopedic medicine. Recent studies showed that piezoelectricity can impact microenvironments by regulating molecular sensors including ion channels, cytoskeletal elements, cell adhesion proteins, and other signaling pathways. This review thus focuses on discussing the pioneering applications of piezoelectricity in the diagnosis and treatment of orthopedic diseases, aiming to offer valuable insights for advancing next-generation medical technologies. Beginning with an introduction to the principles of piezoelectricity and various piezoelectric materials, this review paper delves into the mechanisms through which piezoelectric materials accelerated osteogenesis. A comprehensive overview of piezoelectric materials, devices, and systems enhancing bone tissue repair, alleviating inflammation at infection sites, and monitoring bone health is then provided, respectively. Finally, the major challenges faced by applications of piezoelectricity in orthopedic conditions are thoroughly discussed, along with a critical outlook on future development trends.
Collapse
Affiliation(s)
- Jingkai Zhang
- Department of OrthopaedicsShanghai Key Laboratory for Prevention and Treatment of Bone and Joint DiseasesShanghai Institute of Traumatology and OrthopaedicsRuijin HospitalShanghai Jiao Tong University School of MedicineShanghai200025China
| | - Chang Liu
- Department of OphthalmologyShanghai Ninth People's HospitalShanghai JiaoTong University School of MedicineShanghai200011China
| | - Jun Li
- Department of Materials Science and EngineeringUniversity of Wisconsin–MadisonMadisonWI53706USA
| | - Tao Yu
- Department of OrthopaedicsShanghai Key Laboratory for Prevention and Treatment of Bone and Joint DiseasesShanghai Institute of Traumatology and OrthopaedicsRuijin HospitalShanghai Jiao Tong University School of MedicineShanghai200025China
| | - Jing Ruan
- Department of OphthalmologyShanghai Ninth People's HospitalShanghai JiaoTong University School of MedicineShanghai200011China
| | - Fan Yang
- Department of OrthopaedicsShanghai Key Laboratory for Prevention and Treatment of Bone and Joint DiseasesShanghai Institute of Traumatology and OrthopaedicsRuijin HospitalShanghai Jiao Tong University School of MedicineShanghai200025China
- Research Institute of Frontier ScienceSouthwest Jiaotong UniversityChengduSichuan610031China
| |
Collapse
|
3
|
Ochiai N, Etani Y, Noguchi T, Miura T, Kurihara T, Fukuda Y, Hamada H, Uemura K, Takashima K, Tamaki M, Ishibashi T, Ito S, Yamakawa S, Kanamoto T, Okada S, Nakata K, Ebina K. The pivotal role of the Hes1/Piezo1 pathway in the pathophysiology of glucocorticoid-induced osteoporosis. JCI Insight 2024; 9:e179963. [PMID: 39641269 PMCID: PMC11623955 DOI: 10.1172/jci.insight.179963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Accepted: 10/22/2024] [Indexed: 12/07/2024] Open
Abstract
Glucocorticoid-induced osteoporosis (GIOP) lacks fully effective treatments. This study investigated the role of Piezo1, a mechanosensitive ion channel component 1, in GIOP. We found reduced Piezo1 expression in cortical bone osteocytes from patients with GIOP and a GIOP mouse model. Yoda1, a Piezo1 agonist, enhanced the mechanical stress response and bone mass and strength, which were diminished by dexamethasone (DEX) administration in GIOP mice. RNA-seq revealed that Yoda1 elevated Piezo1 expression by activating the key transcription factor Hes1, followed by enhanced CaM kinase II and Akt phosphorylation in osteocytes. This improved the lacuno-canalicular network and reduced sclerostin production and the receptor activator of NF-κB/osteoprotegerin ratio, which were mitigated by DEX. Comparative analysis of mouse models and human GIOP cortical bone revealed downregulation of mechanostimulated osteogenic factors, such as osteocrin, and cartilage differentiation markers in osteoprogenitor cells. In human periosteum-derived cells, DEX suppressed differentiation into osteoblasts, but Yoda1 rescued this effect. Our findings suggest that reduced Piezo1 expression and activity in osteocytes and periosteal cells contribute to GIOP, and Yoda1 may offer a novel therapeutic approach by restoring mechanosensitivity.
Collapse
Affiliation(s)
- Nagahiro Ochiai
- Department of Musculoskeletal Regenerative Medicine, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
- Taisho Pharmaceutical Co., Ltd., Saitama, Japan
| | - Yuki Etani
- Department of Musculoskeletal Regenerative Medicine, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | | | | | | | | | | | | | | | | | - Teruya Ishibashi
- Department of Orthopaedic Biomaterial Science, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Shohei Ito
- Taisho Pharmaceutical Co., Ltd., Saitama, Japan
| | | | - Takashi Kanamoto
- Department of Health and Sport Sciences, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | | | - Ken Nakata
- Department of Health and Sport Sciences, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Kosuke Ebina
- Department of Musculoskeletal Regenerative Medicine, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
- Department of Orthopaedic Surgery
| |
Collapse
|
4
|
Wasi M, Chu T, Guerra RM, Kooker R, Maldonado K, Li X, Lin CY, Song X, Xiong J, You L, Wang L. Mitigating aging and doxorubicin induced bone loss in mature mice via mechanobiology based treatments. Bone 2024; 188:117235. [PMID: 39147353 PMCID: PMC11475016 DOI: 10.1016/j.bone.2024.117235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 08/08/2024] [Accepted: 08/12/2024] [Indexed: 08/17/2024]
Abstract
Aging leads to a reduced anabolic response to mechanical stimuli and a loss of bone mass and structural integrity. Chemotherapy agents such as doxorubicin exacerbate the degeneration of aging skeleton and further subject older cancer patients to a higher fracture risk. To alleviate this clinical problem, we proposed and tested a novel mechanobiology-based therapy. Building upon prior findings that i) Yoda1, the Piezo1 agonist, promoted bone growth in young adult mice and suppressed bone resorption markers in aged mice, and ii) moderate tibial loading protected bone from breast cancer-induced osteolysis, we hypothesized that combined Yoda1 and moderate loading would improve the structural integrity of adult and aged skeletons in vivo and protect bones from deterioration after chemotherapy. We first examined the effects of 4-week Yoda1 (dose 5 mg/kg, 5 times/week) and moderate tibial loading (4.5 N peak load, 4 Hz, 300 cycles for 5 days/week), individually and combined, on mature mice (∼50 weeks of age). Combined Yoda1 and loading was found to mitigate age-associated cortical and trabecular bone loss better than individual interventions. As expected, the non-treated controls experienced an average drop of cortical polar moment of inertia (Ct.pMOI) by -4.3 % over four weeks and the bone deterioration occurred in the majority (64 %) of the samples. Relative to no treatment, loading alone, Yoda1 alone, and combined Yoda1 and loading increased Ct.pMOI by +7.3 %, +9.5 %, +12.0 % and increased the % of samples with positive Ct.pMOI changes by +32 %, +26 %, and +43 %, respectively, suggesting an additive protection of aging-related bone loss for the combined therapy. We further tested if the treatment efficacy was preserved in mature mice following two weeks (six injections) of doxorubicin at the dose of 2.5 or 5 mg/kg. As expected, doxorubicin increased osteocyte apoptosis, altered bone remodeling, and impaired bone structure. However, the effects induced by DOX were too severe to be rescued by Yoda1 and loading, alone or combined, although loading and Yoda1 individually, or combined, increased the number of mice showing positive responsiveness by 0 %, +15 %, and +29 % relative to no intervention after doxorubicin exposure. Overall, this study supported the potentials and challenges of the Yoda1-based strategy in mitigating the detrimental skeletal effects caused by aging and doxorubicin.
Collapse
Affiliation(s)
- Murtaza Wasi
- Department of Mechanical Engineering, University of Delaware, Newark, DE, USA
| | - Tiankuo Chu
- Department of Mechanical Engineering, University of Delaware, Newark, DE, USA
| | - Rosa M Guerra
- Department of Biomedical Engineering, University of Delaware, Newark, DE, USA
| | - Rory Kooker
- Department of Mechanical Engineering, University of Delaware, Newark, DE, USA
| | - Kenneth Maldonado
- Department of Biomedical Engineering, Kansas State University, Manhattan, KS, USA
| | - Xuehua Li
- Department of Orthopaedic Surgery, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Chun-Yu Lin
- Department of Mechanical and Industrial Engineering, Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
| | - Xin Song
- Department of Mechanical and Industrial Engineering, Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
| | - Jinhu Xiong
- Department of Orthopaedic Surgery, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Lidan You
- Department of Mechanical and Industrial Engineering, Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
| | - Liyun Wang
- Department of Mechanical Engineering, University of Delaware, Newark, DE, USA; Department of Biomedical Engineering, University of Delaware, Newark, DE, USA.
| |
Collapse
|
5
|
Xie B, He X, Guo Y, Shen J, Yang B, Cai R, Chen J, He Y. Cyclic tensile stress promotes osteogenic differentiation via upregulation of Piezo1 in human dental follicle stem cells. Hum Cell 2024; 37:1649-1662. [PMID: 39190266 DOI: 10.1007/s13577-024-01123-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Accepted: 08/17/2024] [Indexed: 08/28/2024]
Abstract
As periodontal progenitor cells, human dental follicle stem cells (hDFCs) play an important role in regenerative medicine research. Mechanical stimuli exert different regulatory effects on various functions of stem cells. Mechanosensitive ion channels can perceive and transmit mechanical signals. Piezo1 is a novel mechanosensitive cation channel dominated by Ca2+ permeation. The yes-associated protein 1 (YAP1) and mitogen-activated protein kinase (MAPK) pathways can respond to mechanical stimuli and play important roles in cell growth, differentiation, apoptosis, and cell cycle regulation. In this study, we demonstrated that Piezo1 was able to transduce cyclic tension stress (CTS) and promote the osteogenic differentiation of hDFCs by applying CTS of 2000 μstrain to hDFCs. Further investigation of this mechanism revealed that CTS activated Piezo1 in hDFCs and resulted in increased levels of intracellular Ca2+, YAP1 nuclear translocation, and phosphorylated protein expression levels of extracellular signalling-associated kinase 1/2 (ERK 1/2) and Jun amino-terminal kinase 1/2/3 (JNK 1/3) of the MAPK pathway family. However, when Piezo1 was knocked down in the hDFCs, all these increases disappeared. We conclude that CTS activates Piezo1 expression and promotes its osteogenesis via Ca2+/YAP1/MAPK in hDFCs. Appropriate mechanical stimulation promotes the osteogenic differentiation of hDFCs via Piezo1. Targeting Piezo1 may be an effective strategy to regulate the osteogenic differentiation of hDFCs, contributing to MSC-based therapies in the field of bone tissue engineering.
Collapse
Affiliation(s)
- Binqing Xie
- Department of Oral and Maxillofacial Surgery, The Affiliated Stomatological Hospital, Southwest Medical University, Yunfenglu 10, Luzhou, 646000, China
- Luzhou Key Laboratory of Oral and Maxillofacial Reconstruction and Regeneration, Southwest Medical University, Xianglinlu 1, Luzhou, 646000, China
| | - Xianyi He
- Department of Oral and Maxillofacial Surgery, The Affiliated Stomatological Hospital, Southwest Medical University, Yunfenglu 10, Luzhou, 646000, China
- Luzhou Key Laboratory of Oral and Maxillofacial Reconstruction and Regeneration, Southwest Medical University, Xianglinlu 1, Luzhou, 646000, China
| | - Ye Guo
- Department of Oral and Maxillofacial Surgery, The Affiliated Stomatological Hospital, Southwest Medical University, Yunfenglu 10, Luzhou, 646000, China
- Luzhou Key Laboratory of Oral and Maxillofacial Reconstruction and Regeneration, Southwest Medical University, Xianglinlu 1, Luzhou, 646000, China
| | - Jie Shen
- Department of Oral and Maxillofacial Surgery, The Affiliated Stomatological Hospital, Southwest Medical University, Yunfenglu 10, Luzhou, 646000, China
- Luzhou Key Laboratory of Oral and Maxillofacial Reconstruction and Regeneration, Southwest Medical University, Xianglinlu 1, Luzhou, 646000, China
| | - Binbin Yang
- Luzhou Key Laboratory of Oral and Maxillofacial Reconstruction and Regeneration, Southwest Medical University, Xianglinlu 1, Luzhou, 646000, China
| | - Rui Cai
- Luzhou Key Laboratory of Oral and Maxillofacial Reconstruction and Regeneration, Southwest Medical University, Xianglinlu 1, Luzhou, 646000, China
| | - Junliang Chen
- Department of Oral and Maxillofacial Surgery, The Affiliated Stomatological Hospital, Southwest Medical University, Yunfenglu 10, Luzhou, 646000, China.
- Luzhou Key Laboratory of Oral and Maxillofacial Reconstruction and Regeneration, Southwest Medical University, Xianglinlu 1, Luzhou, 646000, China.
| | - Yun He
- Department of Oral and Maxillofacial Surgery, The Affiliated Stomatological Hospital, Southwest Medical University, Yunfenglu 10, Luzhou, 646000, China.
- Luzhou Key Laboratory of Oral and Maxillofacial Reconstruction and Regeneration, Southwest Medical University, Xianglinlu 1, Luzhou, 646000, China.
| |
Collapse
|
6
|
Hagan ML, Tuladhar A, Yu K, Alhamad DW, Bensreti H, Dorn J, Piedra VM, Cantu N, Stokes EG, Blumenthal D, Roberts RL, Balayan V, Bass SM, Dickerson T, Cartelle AL, Montesinos-Cartagena M, Awad ME, Castro AA, Garland T, Cooley MA, Johnson M, Hamrick MW, McNeil PL, McGee-Lawrence ME. Osteocyte Sptbn1 Deficiency Alters Cell Survival and Mechanotransduction Following Formation of Plasma Membrane Disruptions (PMD) from Mechanical Loading. Calcif Tissue Int 2024; 115:725-743. [PMID: 39276238 DOI: 10.1007/s00223-024-01285-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Accepted: 08/30/2024] [Indexed: 09/16/2024]
Abstract
We and others have shown that application of high-level mechanical loading promotes the formation of transient plasma membrane disruptions (PMD) which initiate mechanotransduction. We hypothesized that increasing osteocyte cell membrane fragility, by disrupting the cytoskeleton-associated protein β2-spectrin (Sptbn1), could alter osteocytic responses and bone adaptation to loading in a PMD-related fashion. In MLO-Y4 cells, treatment with the spectrin-disrupting agent diamide or knockdown of Sptbn1 via siRNA increased the number of PMD formed by fluid shear stress. Primary osteocytes from an osteocyte-targeted DMP1-Cre Sptbn1 conditional knockout (CKO) model mimicked trends seen with diamide and siRNA treatment and suggested the creation of larger PMD, which repaired more slowly, for a given level of stimulus. Post-wounding cell survival was impaired in all three models, and calcium signaling responses from the wounded osteocyte were mildly altered in Sptbn1 CKO cultures. Although Sptbn1 CKO mice did not demonstrate an altered skeletal phenotype as compared to WT littermates under baseline conditions, they showed a blunted increase in cortical thickness when subjected to an osteogenic tibial loading protocol as well as evidence of increased osteocyte death (increased lacunar vacancy) in the loaded limb after 2 weeks of loading. The impaired post-wounding cell viability and impaired bone adaptation seen with Sptbn1 disruption support the existence of an important role for Sptbn1, and PMD formation, in osteocyte mechanotransduction and bone adaptation to mechanical loading.
Collapse
Affiliation(s)
- Mackenzie L Hagan
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, 1460 Laney Walker Blvd, CB1101, Augusta, GA, 30912, USA
| | - Anik Tuladhar
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, 1460 Laney Walker Blvd, CB1101, Augusta, GA, 30912, USA
| | - Kanglun Yu
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, 1460 Laney Walker Blvd, CB1101, Augusta, GA, 30912, USA
| | - Dima W Alhamad
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, 1460 Laney Walker Blvd, CB1101, Augusta, GA, 30912, USA
| | - Husam Bensreti
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, 1460 Laney Walker Blvd, CB1101, Augusta, GA, 30912, USA
| | - Jennifer Dorn
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, 1460 Laney Walker Blvd, CB1101, Augusta, GA, 30912, USA
| | - Victor M Piedra
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, 1460 Laney Walker Blvd, CB1101, Augusta, GA, 30912, USA
| | - Nicholas Cantu
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, 1460 Laney Walker Blvd, CB1101, Augusta, GA, 30912, USA
| | - Eric G Stokes
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, 1460 Laney Walker Blvd, CB1101, Augusta, GA, 30912, USA
| | - Daniel Blumenthal
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, 1460 Laney Walker Blvd, CB1101, Augusta, GA, 30912, USA
| | - Rachel L Roberts
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, 1460 Laney Walker Blvd, CB1101, Augusta, GA, 30912, USA
| | - Vanshika Balayan
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, 1460 Laney Walker Blvd, CB1101, Augusta, GA, 30912, USA
| | - Sarah M Bass
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, 1460 Laney Walker Blvd, CB1101, Augusta, GA, 30912, USA
| | - Thomas Dickerson
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, 1460 Laney Walker Blvd, CB1101, Augusta, GA, 30912, USA
| | - Anabel Liyen Cartelle
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, 1460 Laney Walker Blvd, CB1101, Augusta, GA, 30912, USA
| | - Marlian Montesinos-Cartagena
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, 1460 Laney Walker Blvd, CB1101, Augusta, GA, 30912, USA
| | - Mohamed E Awad
- Department of Oral Biology and Diagnostic Sciences, Dental College of Georgia, Augusta University, Augusta, GA, USA
| | - Alberto A Castro
- Evolution Ecology & Organismal Biology Department, University of California Riverside, Riverside, USA
| | - Theodore Garland
- Evolution Ecology & Organismal Biology Department, University of California Riverside, Riverside, USA
| | - Marion A Cooley
- Department of Oral Biology and Diagnostic Sciences, Dental College of Georgia, Augusta University, Augusta, GA, USA
| | - Maribeth Johnson
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Mark W Hamrick
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, 1460 Laney Walker Blvd, CB1101, Augusta, GA, 30912, USA
| | - Paul L McNeil
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, 1460 Laney Walker Blvd, CB1101, Augusta, GA, 30912, USA
| | - Meghan E McGee-Lawrence
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, 1460 Laney Walker Blvd, CB1101, Augusta, GA, 30912, USA.
- Department of Orthopaedic Surgery, Augusta University, Augusta, GA, USA.
| |
Collapse
|
7
|
Tuladhar A, Shaver JC, McGee WA, Yu K, Dorn J, Horne JL, Alhamad DW, Hagan ML, Cooley MA, Zhong R, Bollag W, Johnson M, Hamrick MW, McGee-Lawrence ME. Prkd1 regulates the formation and repair of plasma membrane disruptions (PMD) in osteocytes. Bone 2024; 186:117147. [PMID: 38866124 PMCID: PMC11246118 DOI: 10.1016/j.bone.2024.117147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Revised: 05/21/2024] [Accepted: 06/03/2024] [Indexed: 06/14/2024]
Abstract
We and others have seen that osteocytes sense high-impact osteogenic mechanical loading via transient plasma membrane disruptions (PMDs) which initiate downstream mechanotransduction. However, a PMD must be repaired for the cell to survive this wounding event. Previous work suggested that the protein Prkd1 (also known as PKCμ) may be a critical component of this PMD repair process, but the specific role of Prkd1 in osteocyte mechanobiology had not yet been tested. We treated MLO-Y4 osteocytes with Prkd1 inhibitors (Go6976, kbNB 142-70, staurosporine) and generated an osteocyte-targeted (Dmp1-Cre) Prkd1 conditional knockout (CKO) mouse. PMD repair rate was measured via laser wounding and FM1-43 dye uptake, PMD formation and post-wounding survival were assessed via fluid flow shear stress (50 dyn/cm2), and in vitro osteocyte mechanotransduction was assessed via measurement of calcium signaling. To test the role of osteocyte Prkd1 in vivo, Prkd1 CKO and their wildtype (WT) littermates were subjected to 2 weeks of unilateral axial tibial loading and loading-induced changes in cortical bone mineral density, geometry, and formation were measured. Prkd1 inhibition or genetic deletion slowed osteocyte PMD repair rate and impaired post-wounding cell survival. These effects could largely be rescued by treating osteocytes with the FDA-approved synthetic copolymer Poloxamer 188 (P188), which was previously shown to facilitate membrane resealing and improve efficiency in the repair rate of PMD in skeletal muscle myocytes. In vivo, while both WT and Prkd1 CKO mice demonstrated anabolic responses to tibial loading, the magnitude of loading-induced increases in tibial BMD, cortical thickness, and periosteal mineralizing surface were blunted in Prkd1 CKO as compared to WT mice. Prkd1 CKO mice also tended to show a smaller relative difference in the number of osteocyte PMD in loaded limbs and showed greater lacunar vacancy, suggestive of impaired post-wounding osteocyte survival. While P188 treatment rescued loading-induced increases in BMD in the Prkd1 CKO mice, it surprisingly further suppressed loading-induced increases in cortical bone thickness and cortical bone formation. Taken together, these data suggest that Prkd1 may play a pivotal role in the regulation and repair of the PMD response in osteocytes and support the idea that PMD repair processes can be pharmacologically targeted to modulate downstream responses, but suggest limited utility of PMD repair-promoting P188 in improving bone anabolic responses to loading.
Collapse
Affiliation(s)
- Anik Tuladhar
- Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University, Augusta, GA, United States of America
| | - Joseph C Shaver
- Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University, Augusta, GA, United States of America
| | - Wesley A McGee
- Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University, Augusta, GA, United States of America
| | - Kanglun Yu
- Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University, Augusta, GA, United States of America
| | - Jennifer Dorn
- Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University, Augusta, GA, United States of America
| | - J Luke Horne
- Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University, Augusta, GA, United States of America
| | - Dima W Alhamad
- Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University, Augusta, GA, United States of America
| | - Mackenzie L Hagan
- Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University, Augusta, GA, United States of America
| | - Marion A Cooley
- Department of Oral Biology and Diagnostic Sciences, Dental College of Georgia at Augusta University, Augusta, GA, United States of America
| | - Roger Zhong
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia at AugustaUniversity, Augusta, GA, United States of America
| | - Wendy Bollag
- Department of Physiology, Medical College of Georgia at Augusta University, Augusta, GA, United States of America; Charlie Norwood VA Medical Center, Augusta, GA, United States of America
| | - Maribeth Johnson
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia at AugustaUniversity, Augusta, GA, United States of America
| | - Mark W Hamrick
- Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University, Augusta, GA, United States of America
| | - Meghan E McGee-Lawrence
- Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University, Augusta, GA, United States of America.
| |
Collapse
|
8
|
Aragona M, Mhalhel K, Pansera L, Montalbano G, Guerrera MC, Levanti M, Laurà R, Abbate F, Vega JA, Germanà A. Localization of Piezo 1 and Piezo 2 in Lateral Line System and Inner Ear of Zebrafish ( Danio rerio). Int J Mol Sci 2024; 25:9204. [PMID: 39273152 PMCID: PMC11395407 DOI: 10.3390/ijms25179204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 08/20/2024] [Accepted: 08/23/2024] [Indexed: 09/15/2024] Open
Abstract
Piezo proteins have been identified as mechanosensitive ion channels involved in mechanotransduction. Several ion channel dysfunctions may be associated with diseases (including deafness and pain); thus, studying them is critical to understand their role in mechanosensitive disorders and to establish new therapeutic strategies. The current study investigated for the first time the expression patterns of Piezo proteins in zebrafish octavolateralis mechanosensory organs. Piezo 1 and 2 were immunoreactive in the sensory epithelia of the lateral line system and the inner ear. Piezo 1 (28.7 ± 1.55 cells) and Piezo 2 (28.8 ± 3.31 cells) immunopositive neuromast cells were identified based on their ultrastructural features, and their overlapping immunoreactivity to the s100p specific marker (28.6 ± 1.62 cells), as sensory cells. These findings are in favor of Piezo proteins' potential role in sensory cell activation, while their expression on mantle cells reflects their implication in the maintenance and regeneration of the neuromast during cell turnover. In the inner ear, Piezo proteins' colocalization with BDNF introduces their potential implication in neuronal plasticity and regenerative events, typical of zebrafish mechanosensory epithelia. Assessing these proteins in zebrafish could open up new scenarios for the roles of these important ionic membrane channels, for example in treating impairments of sensory systems.
Collapse
Affiliation(s)
- Marialuisa Aragona
- Zebrafish Neuromorphology Lab, Department of Veterinary Sciences, University of Messina, 98168 Messina, Italy
| | - Kamel Mhalhel
- Zebrafish Neuromorphology Lab, Department of Veterinary Sciences, University of Messina, 98168 Messina, Italy
| | - Lidia Pansera
- Zebrafish Neuromorphology Lab, Department of Veterinary Sciences, University of Messina, 98168 Messina, Italy
| | - Giuseppe Montalbano
- Zebrafish Neuromorphology Lab, Department of Veterinary Sciences, University of Messina, 98168 Messina, Italy
| | - Maria Cristina Guerrera
- Zebrafish Neuromorphology Lab, Department of Veterinary Sciences, University of Messina, 98168 Messina, Italy
| | - Maria Levanti
- Zebrafish Neuromorphology Lab, Department of Veterinary Sciences, University of Messina, 98168 Messina, Italy
| | - Rosaria Laurà
- Zebrafish Neuromorphology Lab, Department of Veterinary Sciences, University of Messina, 98168 Messina, Italy
| | - Francesco Abbate
- Zebrafish Neuromorphology Lab, Department of Veterinary Sciences, University of Messina, 98168 Messina, Italy
| | - José A Vega
- Departamento de Morfología y Biología Celular, Grupo SINPOS, Universidad de Oviedo, 33006 Oviedo, Spain
- Facultad de Ciencias de la Salud, Universidad Autónoma de Chile, Santiago 7500912, Chile
| | - Antonino Germanà
- Zebrafish Neuromorphology Lab, Department of Veterinary Sciences, University of Messina, 98168 Messina, Italy
| |
Collapse
|
9
|
Hao Y, Yang N, Sun M, Yang S, Chen X. The role of calcium channels in osteoporosis and their therapeutic potential. Front Endocrinol (Lausanne) 2024; 15:1450328. [PMID: 39170742 PMCID: PMC11335502 DOI: 10.3389/fendo.2024.1450328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Accepted: 07/25/2024] [Indexed: 08/23/2024] Open
Abstract
Osteoporosis, a systemic skeletal disorder marked by diminished bone mass and compromised bone microarchitecture, is becoming increasingly prevalent due to an aging population. The underlying pathophysiology of osteoporosis is attributed to an imbalance between osteoclast-mediated bone resorption and osteoblast-mediated bone formation. Osteoclasts play a crucial role in the development of osteoporosis through various molecular pathways, including the RANK/RANKL/OPG signaling axis, cytokines, and integrins. Notably, the calcium signaling pathway is pivotal in regulating osteoclast activation and function, influencing bone resorption activity. Disruption in calcium signaling can lead to increased osteoclast-mediated bone resorption, contributing to the progression of osteoporosis. Emerging research indicates that calcium-permeable channels on the cellular membrane play a critical role in bone metabolism by modulating these intracellular calcium pathways. Here, we provide an overview of current literature on the regulation of plasma membrane calcium channels in relation to bone metabolism with particular emphasis on their dysregulation during the progression of osteoporosis. Targeting these calcium channels may represent a potential therapeutic strategy for treating osteoporosis.
Collapse
Affiliation(s)
- Ying Hao
- College of Sports, Northwest Normal University, Lanzhou, China
| | - Ningning Yang
- College of Sports, Northwest Normal University, Lanzhou, China
| | - Mengying Sun
- College of Sports, Northwest Normal University, Lanzhou, China
| | - Shangze Yang
- Institute of Medical Research, Northwestern Polytechnical University, Xi’an, China
| | - Xingjuan Chen
- Institute of Medical Research, Northwestern Polytechnical University, Xi’an, China
| |
Collapse
|
10
|
Ubuzima P, Nshimiyimana E, Mukeshimana C, Mazimpaka P, Mugabo E, Mbyayingabo D, Mohamed AS, Habumugisha J. Exploring biological mechanisms in orthodontic tooth movement: Bridging the gap between basic research experiments and clinical applications - A comprehensive review. Ann Anat 2024; 255:152286. [PMID: 38810763 DOI: 10.1016/j.aanat.2024.152286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 04/21/2024] [Accepted: 05/21/2024] [Indexed: 05/31/2024]
Abstract
OBJECTIVES The molecular mechanisms behind orthodontic tooth movements (OTM) were investigated by clarifying the role of chemical messengers released by cells. METHODS Using the Cochrane library, Google scholar, and PubMed databases, a literature search was conducted, and studies published from 1984 to 2024 were considered. RESULTS Both bone growth and remodeling may occur when a tooth is subjected to mechanical stress. These chemicals have a significant effect on the stimulation and regulation of osteoblasts, osteoclasts, and osteocytes during alveolar bone remodeling. This regulation can take place in pathological conditions, such as periodontal diseases, or during OTM alone. This comprehensive review outlines key molecular mechanisms underlying OTM and explores various clinical assumptions associated with specific molecules and their functional domains during this process. Furthermore, clinical applications of certain molecules such as relaxin, prostaglandin E (PGE), and interleukin-1β (IL-1β) in accelerating OTM have been reported. Our findings underscore the existing gap between OTM clinical applications and basic research investigations. CONCLUSION A comprehensive understanding of orthodontic treatment is enriched by insights into biological systems. We reported the activation of osteoblasts, osteoclast precursor cells, osteoclasts, and osteocytes in response to mechanical stress, leading to targeted cellular and molecular interventions and facilitating rapid and regulated alveolar bone remodeling during tooth movement. Despite the shortcomings of clinical studies in accelerating OTM, this review highlights the crucial role of biological agents in this process and advocates for prioritizing high-quality human studies in future research to gain further insights from clinical trials.
Collapse
Affiliation(s)
- Pascal Ubuzima
- Department of Orthodontics, Affliated Hospital of Stomatology, Anhui Medical University Hefei, 69 Meishan Road, Hefei, Anhui, China; School of Dentistry, College of Medicine and Health Sciences, University of Rwanda, Rwanda
| | - Eugene Nshimiyimana
- Department of Orthodontics, Affliated Hospital of Stomatology, Anhui Medical University Hefei, 69 Meishan Road, Hefei, Anhui, China
| | - Christelle Mukeshimana
- Department of Orthodontics, Affliated Hospital of Stomatology, Anhui Medical University Hefei, 69 Meishan Road, Hefei, Anhui, China
| | - Patrick Mazimpaka
- School of Dentistry, College of Medicine and Health Sciences, University of Rwanda, Rwanda
| | - Eric Mugabo
- Department of Orthodontics, Xiangya Stomatological Hospital & Xiangya School of Stomatology, Central South University, 72 Xiangya Road, Changsha, Hunan 410000, China
| | - Dieudonne Mbyayingabo
- Department of Orthodontics, Stomatological Hospital of Xi'an Jiaotong University, 98 XiWu Road, Xi'an, Shaanxi 710004, China
| | | | - Janvier Habumugisha
- Department of Orthodontics, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, 2-5-1, Shikata-cho, Kitaku, Okayama 700-8525, Japan; Department of Biochemistry and Molecular Dentistry, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan.
| |
Collapse
|
11
|
Wu Z, Li W, Jiang K, Lin Z, Qian C, Wu M, Xia Y, Li N, Zhang H, Xiao H, Bai J, Geng D. Regulation of bone homeostasis: signaling pathways and therapeutic targets. MedComm (Beijing) 2024; 5:e657. [PMID: 39049966 PMCID: PMC11266958 DOI: 10.1002/mco2.657] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 06/22/2024] [Accepted: 06/25/2024] [Indexed: 07/27/2024] Open
Abstract
As a highly dynamic tissue, bone is continuously rebuilt throughout life. Both bone formation by osteoblasts and bone resorption by osteoclasts constitute bone reconstruction homeostasis. The equilibrium of bone homeostasis is governed by many complicated signaling pathways that weave together to form an intricate network. These pathways coordinate the meticulous processes of bone formation and resorption, ensuring the structural integrity and dynamic vitality of the skeletal system. Dysregulation of the bone homeostatic regulatory signaling network contributes to the development and progression of many skeletal diseases. Significantly, imbalanced bone homeostasis further disrupts the signaling network and triggers a cascade reaction that exacerbates disease progression and engenders a deleterious cycle. Here, we summarize the influence of signaling pathways on bone homeostasis, elucidating the interplay and crosstalk among them. Additionally, we review the mechanisms underpinning bone homeostatic imbalances across diverse disease landscapes, highlighting current and prospective therapeutic targets and clinical drugs. We hope that this review will contribute to a holistic understanding of the signaling pathways and molecular mechanisms sustaining bone homeostasis, which are promising to contribute to further research on bone homeostasis and shed light on the development of targeted drugs.
Collapse
Affiliation(s)
- Zebin Wu
- Department of OrthopedicsThe First Affiliated Hospital of Soochow UniversitySuzhouJiangsuChina
| | - Wenming Li
- Department of OrthopedicsThe First Affiliated Hospital of Soochow UniversitySuzhouJiangsuChina
| | - Kunlong Jiang
- Department of OrthopedicsThe First Affiliated Hospital of Soochow UniversitySuzhouJiangsuChina
| | - Zhixiang Lin
- Department of OrthopedicsThe First Affiliated Hospital of Soochow UniversitySuzhouJiangsuChina
| | - Chen Qian
- Department of OrthopedicsThe First Affiliated Hospital of Soochow UniversitySuzhouJiangsuChina
| | - Mingzhou Wu
- Department of OrthopedicsThe First Affiliated Hospital of Soochow UniversitySuzhouJiangsuChina
| | - Yu Xia
- Department of OrthopedicsThe First Affiliated Hospital of Soochow UniversitySuzhouJiangsuChina
| | - Ning Li
- Department of OrthopedicsCentre for Leading Medicine and Advanced Technologies of IHMDivision of Life Sciences and MedicineThe First Affiliated Hospital of USTCUniversity of Science and Technology of ChinaHefeiChina
| | - Hongtao Zhang
- Department of OrthopedicsThe First Affiliated Hospital of Soochow UniversitySuzhouJiangsuChina
| | - Haixiang Xiao
- Department of OrthopedicsThe First Affiliated Hospital of Soochow UniversitySuzhouJiangsuChina
- Department of OrthopedicsJingjiang People's HospitalSeventh Clinical Medical School of Yangzhou UniversityJingjiangJiangsu ProvinceChina
| | - Jiaxiang Bai
- Department of OrthopedicsCentre for Leading Medicine and Advanced Technologies of IHMDivision of Life Sciences and MedicineThe First Affiliated Hospital of USTCUniversity of Science and Technology of ChinaHefeiChina
| | - Dechun Geng
- Department of OrthopedicsThe First Affiliated Hospital of Soochow UniversitySuzhouJiangsuChina
| |
Collapse
|
12
|
Thien ND, Hai-Nam N, Anh DT, Baecker D. Piezo1 and its inhibitors: Overview and perspectives. Eur J Med Chem 2024; 273:116502. [PMID: 38761789 DOI: 10.1016/j.ejmech.2024.116502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Revised: 05/11/2024] [Accepted: 05/13/2024] [Indexed: 05/20/2024]
Abstract
The cation channel Piezo1, a crucial mechanotransducer found in various organs and tissues, has gained considerable attention as a therapeutic target in recent years. Following this trend, several Piezo1 inhibitors have been discovered and studied for potential pharmacological properties. This review provides an overview of the structural and functional importance of Piezo1, as well as discussing the biological activities of Piezo1 inhibitors based on their mechanism of action. The compounds addressed include the toxin GsMTx4, Aβ peptides, certain fatty acids, ruthenium red and gadolinium, Dooku1, as well as the natural products tubeimoside I, salvianolic acid B, jatrorrhzine, and escin. The findings revealed that misexpression of Piezo1 can be associated with a number of chronic diseases, including hypertension, cancer, and hemolytic anemia. Consequently, inhibiting Piezo1 and the subsequent calcium influx can have beneficial effects on various pathological processes, as shown by many in vitro and in vivo studies. However, the development of Piezo1 inhibitors is still in its beginnings, with many opportunities and challenges remaining to be explored.
Collapse
Affiliation(s)
- Nguyen Duc Thien
- Hanoi University of Pharmacy, 13-15 Le Thanh Tong, Hanoi, 100000, Viet Nam
| | - Nguyen Hai-Nam
- Hanoi University of Pharmacy, 13-15 Le Thanh Tong, Hanoi, 100000, Viet Nam
| | - Duong Tien Anh
- Hanoi University of Pharmacy, 13-15 Le Thanh Tong, Hanoi, 100000, Viet Nam.
| | - Daniel Baecker
- Department of Pharmaceutical and Medicinal Chemistry, Institute of Pharmacy, Freie Universität Berlin, Königin-Luise-Straße 2+4, Berlin, 14195, Germany.
| |
Collapse
|
13
|
Owen R, Wittkowske C, Lacroix D, Perrault CM, Reilly GC. β-glycerophosphate, not low magnitude fluid shear stress, increases osteocytogenesis in the osteoblast-to-osteocyte cell line IDG-SW3. Connect Tissue Res 2024; 65:313-329. [PMID: 38982804 PMCID: PMC11371265 DOI: 10.1080/03008207.2024.2375065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 06/25/2024] [Accepted: 06/26/2024] [Indexed: 07/11/2024]
Abstract
AIM As osteoblasts deposit a mineralized collagen network, a subpopulation of these cells differentiates into osteocytes. Biochemical and mechanical stimuli, particularly fluid shear stress (FSS), are thought to regulate this, but their relative influence remains unclear. Here, we assess both biochemical and mechanical stimuli on long-term bone formation and osteocytogenesis using the osteoblast-osteocyte cell line IDG-SW3. METHODS Due to the relative novelty and uncommon culture conditions of IDG-SW3 versus other osteoblast-lineage cell lines, effects of temperature and media formulation on matrix deposition and osteocytogenesis were initially characterized. Subsequently, the relative influence of biochemical (β-glycerophosphate (βGP) and ascorbic acid 2-phosphate (AA2P)) and mechanical stimulation on osteocytogenesis was compared, with intermittent application of low magnitude FSS generated by see-saw rocker. RESULTS βGP and AA2P supplementation were required for mineralization and osteocytogenesis, with 33°C cultures retaining a more osteoblastic phenotype and 37°C cultures undergoing significantly higher osteocytogenesis. βGP concentration positively correlated with calcium deposition, whilst AA2P stimulated alkaline phosphatase (ALP) activity and collagen deposition. We demonstrate that increasing βGP concentration also significantly enhances osteocytogenesis as quantified by the expression of green fluorescent protein linked to Dmp1. Intermittent FSS (~0.06 Pa) rocker had no effect on osteocytogenesis and matrix deposition. CONCLUSIONS This work demonstrates the suitability and ease with which IDG-SW3 can be utilized in osteocytogenesis studies. IDG-SW3 mineralization was only mediated through biochemical stimuli with no detectable effect of low magnitude FSS. Osteocytogenesis of IDG-SW3 primarily occurred in mineralized areas, further demonstrating the role mineralization of the bone extracellular matrix has in osteocyte differentiation.
Collapse
Affiliation(s)
- Robert Owen
- Department of Materials Science and Engineering, University of Sheffield, Sheffield, UK
- INSIGNEO Institute for In Silico Medicine, University of Sheffield, Sheffield, UK
- School of Pharmacy, University of Nottingham Biodiscovery Institute, University of Nottingham, Nottingham, UK
| | - Claudia Wittkowske
- INSIGNEO Institute for In Silico Medicine, University of Sheffield, Sheffield, UK
- Department of Mechanical Engineering, University of Sheffield, Sheffield, UK
| | - Damien Lacroix
- INSIGNEO Institute for In Silico Medicine, University of Sheffield, Sheffield, UK
- Department of Mechanical Engineering, University of Sheffield, Sheffield, UK
| | - Cecile M. Perrault
- INSIGNEO Institute for In Silico Medicine, University of Sheffield, Sheffield, UK
- Department of Mechanical Engineering, University of Sheffield, Sheffield, UK
| | - Gwendolen C. Reilly
- Department of Materials Science and Engineering, University of Sheffield, Sheffield, UK
- INSIGNEO Institute for In Silico Medicine, University of Sheffield, Sheffield, UK
| |
Collapse
|
14
|
Kang T, Yang Z, Zhou M, Lan Y, Hong Y, Gong X, Wu Y, Li M, Chen X, Zhang W. The role of the Piezo1 channel in osteoblasts under cyclic stretching: A study on osteogenic and osteoclast factors. Arch Oral Biol 2024; 163:105963. [PMID: 38608563 DOI: 10.1016/j.archoralbio.2024.105963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 03/10/2024] [Accepted: 03/25/2024] [Indexed: 04/14/2024]
Abstract
OBJECTIVES Orthodontic tooth movement is a mechanobiological reaction induced by appropriate forces, including bone remodeling. The mechanosensitive Piezo channels have been shown to contribute to bone remodeling. However, information about the pathways through which Piezo channels affects osteoblasts remains limited. Thus, we aimed to investigate the influence of Piezo1 on the osteogenic and osteoclast factors in osteoblasts under mechanical load. MATERIALS AND METHODS Cyclic stretch (CS) experiments on MC3T3-E1 were conducted using a BioDynamic mechanical stretching device. The Piezo1 channel blocker GsMTx4 and the Piezo1 channel agonist Yoda1 were used 12 h before the application of CS. MC3T3-E1 cells were then subjected to 15% CS, and the expression of Piezo1, Piezo2, BMP-2, OCN, Runx2, RANKL, p-p65/p65, and ALP was measured using quantitative real-time polymerase chain reaction, western blot, alkaline phosphatase staining, and immunofluorescence staining. RESULTS CS of 15% induced the highest expression of Piezo channel and osteoblast factors. Yoda1 significantly increased the CS-upregulated expression of Piezo1 and ALP activity but not Piezo2 and RANKL. GsMTx4 downregulated the CS-upregulated expression of Piezo1, Piezo2, Runx2, OCN, p-65/65, and ALP activity but could not completely reduce CS-upregulated BMP-2. CONCLUSIONS The appropriate force is more suitable for promoting osteogenic differentiation in MC3T3-E1. The Piezo1 channel participates in osteogenic differentiation of osteoblasts through its influence on the expression of osteogenic factors like BMP-2, Runx2, and OCN and is involved in regulating osteoclasts by influencing phosphorylated p65. These results provide a foundation for further exploration of osteoblast function in orthodontic tooth movement.
Collapse
Affiliation(s)
- Ting Kang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, China
| | - Ziyuan Yang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, China
| | - Mengqi Zhou
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, China
| | - Yanhua Lan
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, China
| | - Yaya Hong
- Center for Plastic & Reconstructive Surgery, Department of Stomatology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, China
| | - Xinyi Gong
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, China
| | - Yongjia Wu
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, China
| | - Min Li
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hangzhou, China
| | - Xuepeng Chen
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, China.
| | - Weifang Zhang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, China.
| |
Collapse
|
15
|
Hiasa M, Endo I, Matsumoto T. Bone-fat linkage via interleukin-11 in response to mechanical loading. J Bone Miner Metab 2024; 42:447-454. [PMID: 38324177 DOI: 10.1007/s00774-023-01493-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Accepted: 12/10/2023] [Indexed: 02/08/2024]
Abstract
Positive regulators of bone formation, such as mechanical loading and PTH, stimulate and negative regulators, such as aging and glucocorticoid excess, suppress IL-11 gene transcription in osteoblastic cells. Signal transduction from mechanical loading and PTH stimulation involves two pathways: one is Ca2+-ERK-CREB pathway which facilitates binding of ∆FosB/JunD to the AP-1 site to enhance IL-11 gene transcription, and the other is Smad1/5 phosphorylation that promotes IL-11 gene transcription via SBE binding and complex formation with ∆FosB/JunD. The increased IL-11 suppresses Sost expression via IL-11Rα-STAT1/3-HDAC4/5 pathway and enhances Wnt signaling in the bone to stimulate bone formation. Thus, IL-11 mediates stimulatory and inhibitory signals of bone formation by affecting Wnt signaling. Physiologically important stimulation of bone formation is exercise-induced mechanical loading, but exercise simultaneously requires energy source for muscle contraction. Exercise-induced stimulation of IL-11 expression in the bone increases the secretion of IL-11 from the bone. The increased circulating IL-11 acts like a hormone to enhance adipolysis as an energy source with a reduction in adipogenic differentiation via a suppression of Dkk1/2 expression in the adipose tissue. Such bone-fat linkage can be a mechanism whereby exercise increases bone mass and, at the same time, maintains energy supply from the adipose tissue.
Collapse
Affiliation(s)
- Masahiro Hiasa
- Department of Orthodontics and Dentofacial Orthopedics, Tokushima University Graduate School of Dentistry, Tokushima, 770-8503, Japan
| | - Itsuro Endo
- Department of Endocrinology, Metabolism and Hematology, Tokushima University Graduate School of Medical Sciences, 3-18-15 Kuramoto-Cho, Tokushima, 770-8503,, Japan
| | - Toshio Matsumoto
- Department of Endocrinology, Metabolism and Hematology, Tokushima University Graduate School of Medical Sciences, 3-18-15 Kuramoto-Cho, Tokushima, 770-8503,, Japan.
| |
Collapse
|
16
|
Wang Y, Jiang T. Recent research advances in pain mechanisms in McCune-Albright syndrome thinking about the pain mechanism of FD/MAS. J Orthop Surg Res 2024; 19:196. [PMID: 38515135 PMCID: PMC10956191 DOI: 10.1186/s13018-024-04687-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Accepted: 03/16/2024] [Indexed: 03/23/2024] Open
Abstract
BACKGROUND The lack of effective understanding of the pain mechanism of McCune-Albright syndrome (MAS) has made the treatment of pain in this disease a difficult clinical challenge, and new therapeutic targets are urgently needed to address this dilemma. OBJECTIVE This paper summarizes the novel mechanisms, targets, and treatments that may produce pain in MAS and fibrous dysplasia (polyfibrous dysplasia, or FD). METHODS We conducted a systematic search in the PubMed database, Web of Science, China Knowledge Network (CNKI) with the following keywords: "McCune-Albright syndrome (MAS); polyfibrous dysplasia (FD); bone pain; bone remodeling; G protein coupled receptors; GDNF family receptors; purinergic receptors and glycogen synthase kinase", as well as other keywords were systematically searched. Papers published between January 2018 and May 2023 were selected for finding. Initial screening was performed by reading the titles and abstracts, and available literature was screened against the inclusion and exclusion criteria. RESULTS In this review, we systematically analyzed the cutting-edge advances in this disease, synthesized the findings, and discussed the differences. With regard to the complete mechanistic understanding of the pain condition in FD/MAS, in particular, we collated new findings on new pathways, neurotrophic factor receptors, purinergic receptors, interferon-stimulating factors, potassium channels, protein kinases, and corresponding hormonal modulation and their respective strengths and weaknesses. CONCLUSION This paper focuses on basic research to explore FD/MAS pain mechanisms. New nonneuronal and molecular mechanisms, mechanically loaded responsive neurons, and new targets for potential clinical interventions are future research directions, and a large number of animal experiments, tissue engineering techniques, and clinical trials are still needed to verify the effectiveness of the targets in the future.
Collapse
Affiliation(s)
- Yong Wang
- Orthopedics Department, Changzhou Traditional Chinese Medicine Hospital, Nanjing University of Chinese Medicine, Changzhou, 213000, Jangsu Province, China
| | - Tao Jiang
- Orthopedics Department, Changzhou Traditional Chinese Medicine Hospital, Nanjing University of Chinese Medicine, Changzhou, 213000, Jangsu Province, China.
| |
Collapse
|
17
|
Ogino S, Yoshikawa K, Nagase T, Mikami K, Nagase M. Roles of the mechanosensitive ion channel Piezo1 in the renal podocyte injury of experimental hypertensive nephropathy. Hypertens Res 2024; 47:747-759. [PMID: 38145990 DOI: 10.1038/s41440-023-01536-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2023] [Revised: 11/10/2023] [Accepted: 11/18/2023] [Indexed: 12/27/2023]
Abstract
Glomerular podocyte injury plays an essential role in proteinuria pathogenesis, a hallmark of chronic kidney disease, including hypertensive nephropathy. Although podocytes are susceptible to mechanical stimuli, their mechanotransduction pathways remain elusive. Piezo proteins, including Piezo1 and 2, are mechanosensing ion channels that mediate various biological phenomena. Although renal Piezo2 expression and its alteration in rodent dehydration and hypertension models have been reported, the role of Piezo1 in hypertensive nephropathy and podocyte injury is unclear. In this study, we examined Piezo1 expression and localization in the kidneys of control mice and in those of mice with hypertensive nephrosclerosis. Uninephrectomized, aldosterone-infused, salt-loaded mice developed hypertension, albuminuria, podocyte injury, and glomerulosclerosis. RNAscope in situ hybridization revealed that Piezo1 expression was enhanced in the podocytes, mesangial cells, and distal tubular cells of these mice compared to those of the uninephrectomized, vehicle-infused control group. Piezo1 upregulation in the glomeruli was accompanied by the induction of podocyte injury-related markers, plasminogen activator inhibitor-1 and serum/glucocorticoid regulated kinase 1. These changes were reversed by antihypertensive drug. Exposure of Piezo1-expressing cultured podocytes to mechanical stretch activated Rac1 and upregulated the above-mentioned markers, which was antagonized by the Piezo1 blocker grammostola mechanotoxin #4 (GsMTx4). Administration of Piezo1-specific agonist Yoda1 mimicked the effects of mechanical stretch, which was minimized by the Yoda1-specific inhibitor Dooku1 and Rac inhibitor. Rac1 was also activated in the above-mentioned hypertensive mice, and Rac inhibitor downregulated gene expression of podocyte injury-related markers in vivo. Our results suggest that Piezo1 plays a role in mechanical stress-induced podocyte injury.
Collapse
Affiliation(s)
- Satoyuki Ogino
- Department of Anatomy, Kyorin University School of Medicine, Mitaka, Japan
- Department of Trauma and Critical Care Medicine, Kyorin University School of Medicine, Mitaka, Japan
| | - Kei Yoshikawa
- Department of Anatomy, Kyorin University School of Medicine, Mitaka, Japan
- Department of Trauma and Critical Care Medicine, Kyorin University School of Medicine, Mitaka, Japan
| | - Takashi Nagase
- Kunitachi Aoyagien Tachikawa Geriatric Health Services Facility, Tachikawa, Japan
| | - Kaori Mikami
- Department of Anatomy, Kyorin University School of Medicine, Mitaka, Japan
| | - Miki Nagase
- Department of Anatomy, Kyorin University School of Medicine, Mitaka, Japan.
| |
Collapse
|
18
|
Du Y, Xu B, Li Q, Peng C, Yang K. The role of mechanically sensitive ion channel Piezo1 in bone remodeling. Front Bioeng Biotechnol 2024; 12:1342149. [PMID: 38390363 PMCID: PMC10882629 DOI: 10.3389/fbioe.2024.1342149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Accepted: 01/16/2024] [Indexed: 02/24/2024] Open
Abstract
Piezo1 (2010) was identified as a mechanically activated cation channel capable of sensing various physical forces, such as tension, osmotic pressure, and shear force. Piezo1 mediates mechanosensory transduction in different organs and tissues, including its role in maintaining bone homeostasis. This review aimed to summarize the function and possible mechanism of Piezo1 in the mechanical receptor cells in bone tissue. We found that it is a potential therapeutic target for the treatment of bone diseases.
Collapse
Affiliation(s)
| | | | | | | | - Kai Yang
- Department of Orthodontics, School of Stomatology, Capital Medical University, Beijing, China
| |
Collapse
|
19
|
Inoue S, Li C, Hatakeyama J, Jiang H, Kuroki H, Moriyama H. Higher-intensity ultrasound accelerates fracture healing via mechanosensitive ion channel Piezo1. Bone 2023; 177:116916. [PMID: 37777037 DOI: 10.1016/j.bone.2023.116916] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 09/08/2023] [Accepted: 09/18/2023] [Indexed: 10/02/2023]
Abstract
Osteoporosis-related fractures are a major public health problem. Mechanobiological stimulation utilizing low-intensity pulsed ultrasound (LIPUS) is the most widely accepted modality for accelerating fracture healing. However, recent evidence has demonstrated the ineffectiveness of LIPUS, and the biophysical mechanisms of ultrasound-induced bone formation also remain elusive. Here, we demonstrate that ultrasound at a higher intensity than LIPUS effectively accelerates fracture healing in a mouse osteoporotic fracture model. Higher-intensity ultrasound promoted chondrogenesis and hypertrophic differentiation of chondrocytes in the fracture callus. Higher-intensity ultrasound also increased osteoblasts and newly formed bone in the callus, resulting in accelerated endochondral ossification during fracture healing. In addition, we found that accelerated fracture healing by ultrasound exposure was attenuated when the mechanosensitive ion channel Piezo1 was inhibited by GsMTx4. Ultrasound-induced new bone formation in the callus was attenuated in fractured mice treated with GsMTx4. Similar results were also confirmed in a 3D osteocyte-osteoblast co-culture system, where osteocytic Piezo1 knockdown attenuated the expression of osteoblastic genes after ultrasound exposure. Together these results demonstrate that higher-intensity ultrasound than clinically used LIPUS can accelerate endochondral ossification after fractures. Furthermore, our results suggest that mechanotransduction via Piezo1 mediates ultrasound-stimulated fracture healing and bone formation.
Collapse
Affiliation(s)
- Shota Inoue
- Department of Rehabilitation Science, Graduate School of Health Sciences, Kobe University, Kobe, Japan
| | - Changxin Li
- Department of Rehabilitation Science, Graduate School of Health Sciences, Kobe University, Kobe, Japan
| | - Junpei Hatakeyama
- Department of Rehabilitation Science, Graduate School of Health Sciences, Kobe University, Kobe, Japan; Research Fellowship of the Japan Society for the Promotion of Science, Japan
| | - Hanlin Jiang
- Department of Rehabilitation Science, Graduate School of Health Sciences, Kobe University, Kobe, Japan
| | - Hiroshi Kuroki
- Department of Physical Therapy, Human Health Sciences, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Hideki Moriyama
- Life and Medical Sciences Area, Health Sciences Discipline, Kobe University, Kobe, Japan.
| |
Collapse
|
20
|
Kitase Y, Prideaux M. Regulation of the Osteocyte Secretome with Aging and Disease. Calcif Tissue Int 2023; 113:48-67. [PMID: 37148298 DOI: 10.1007/s00223-023-01089-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Accepted: 04/21/2023] [Indexed: 05/08/2023]
Abstract
As the most numerous and long-lived of all bone cells, osteocytes have essential functions in regulating skeletal health. Through the lacunar-canalicular system, secreted proteins from osteocytes can reach cells throughout the bone. Furthermore, the intimate connectivity between the lacunar-canalicular system and the bone vasculature allows for the transport of osteocyte-secreted factors into the circulation to reach the entire body. Local and endocrine osteocyte signaling regulates physiological processes such as bone remodeling, bone mechanoadaptation, and mineral homeostasis. However, these processes are disrupted by impaired osteocyte function induced by aging and disease. Dysfunctional osteocyte signaling is now associated with the pathogenesis of many disorders, including chronic kidney disease, cancer, diabetes mellitus, and periodontitis. In this review, we focus on the targeting of bone and extraskeletal tissues by the osteocyte secretome. In particular, we highlight the secreted osteocyte proteins, which are known to be dysregulated during aging and disease, and their roles during disease progression. We also discuss how therapeutic or genetic targeting of osteocyte-secreted proteins can improve both skeletal and systemic health.
Collapse
Affiliation(s)
- Yukiko Kitase
- Indiana Center for Musculoskeletal Health, School of Medicine, Indiana University, Indianapolis, IN, 46202, USA
- Department of Anatomy, Cell Biology and Physiology, School of Medicine, Indiana University, Indianapolis, IN, 46202, USA
| | - Matthew Prideaux
- Indiana Center for Musculoskeletal Health, School of Medicine, Indiana University, Indianapolis, IN, 46202, USA.
- Department of Anatomy, Cell Biology and Physiology, School of Medicine, Indiana University, Indianapolis, IN, 46202, USA.
| |
Collapse
|
21
|
Zhang Y, Wang L, Kang H, Lin CY, Fan Y. Unlocking the Therapeutic Potential of Irisin: Harnessing Its Function in Degenerative Disorders and Tissue Regeneration. Int J Mol Sci 2023; 24:ijms24076551. [PMID: 37047523 PMCID: PMC10095399 DOI: 10.3390/ijms24076551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 03/26/2023] [Accepted: 03/28/2023] [Indexed: 04/03/2023] Open
Abstract
Physical activity is well-established as an important protective factor against degenerative conditions and a promoter of tissue growth and renewal. The discovery of Fibronectin domain-containing protein 5 (FNDC5) as the precursor of Irisin in 2012 sparked significant interest in its potential as a diagnostic biomarker and a therapeutic agent for various diseases. Clinical studies have examined the correlation between plasma Irisin levels and pathological conditions using a range of assays, but the lack of reliable measurements for endogenous Irisin has led to uncertainty about its prognostic/diagnostic potential as an exercise surrogate. Animal and tissue-engineering models have shown the protective effects of Irisin treatment in reversing functional impairment and potentially permanent damage, but dosage ambiguities remain unresolved. This review provides a comprehensive examination of the clinical and basic studies of Irisin in the context of degenerative conditions and explores its potential as a therapeutic approach in the physiological processes involved in tissue repair/regeneration.
Collapse
Affiliation(s)
- Yuwei Zhang
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Centre for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, China
| | - Lizhen Wang
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Centre for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, China
- Correspondence:
| | - Hongyan Kang
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Centre for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, China
| | - Chia-Ying Lin
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Centre for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, China
- Department of Biomedical, Chemical & Environmental Engineering, University of Cincinnati, Cincinnati, OH 45267, USA
- Department of Orthopaedic Surgery, University of Cincinnati, Cincinnati, OH 45267, USA
- Department of Neurosurgery, University of Cincinnati, Cincinnati, OH 45267, USA
| | - Yubo Fan
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Centre for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, China
- School of Engineering Medicine, Beihang University, Beijing 100083, China
| |
Collapse
|
22
|
The Impact of Plasma Membrane Ion Channels on Bone Remodeling in Response to Mechanical Stress, Oxidative Imbalance, and Acidosis. Antioxidants (Basel) 2023; 12:antiox12030689. [PMID: 36978936 PMCID: PMC10045377 DOI: 10.3390/antiox12030689] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 03/01/2023] [Accepted: 03/03/2023] [Indexed: 03/14/2023] Open
Abstract
The extracellular milieu is a rich source of different stimuli and stressors. Some of them depend on the chemical–physical features of the matrix, while others may come from the ‘outer’ environment, as in the case of mechanical loading applied on the bones. In addition to these forces, a plethora of chemical signals drives cell physiology and fate, possibly leading to dysfunctions when the homeostasis is disrupted. This variety of stimuli triggers different responses among the tissues: bones represent a particular milieu in which a fragile balance between mechanical and metabolic demands should be tuned and maintained by the concerted activity of cell biomolecules located at the interface between external and internal environments. Plasma membrane ion channels can be viewed as multifunctional protein machines that act as rapid and selective dual-nature hubs, sensors, and transducers. Here we focus on some multisensory ion channels (belonging to Piezo, TRP, ASIC/EnaC, P2XR, Connexin, and Pannexin families) actually or potentially playing a significant role in bone adaptation to three main stressors, mechanical forces, oxidative stress, and acidosis, through their effects on bone cells including mesenchymal stem cells, osteoblasts, osteoclasts, and osteocytes. Ion channel-mediated bone remodeling occurs in physiological processes, aging, and human diseases such as osteoporosis, cancer, and traumatic events.
Collapse
|
23
|
Kalyanaraman H, China SP, Cabriales JA, Moininazeri J, Casteel DE, Garcia JJ, Wong VW, Chen A, Sah RL, Boss GR, Pilz RB. Protein Kinase G2 Is Essential for Skeletal Homeostasis and Adaptation to Mechanical Loading in Male but Not Female Mice. J Bone Miner Res 2023; 38:171-185. [PMID: 36371651 PMCID: PMC9825661 DOI: 10.1002/jbmr.4746] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2022] [Revised: 11/04/2022] [Accepted: 11/10/2022] [Indexed: 11/14/2022]
Abstract
We previously showed that the NO/cGMP/protein kinase G (PKG) signaling pathway positively regulates osteoblast proliferation, differentiation, and survival in vitro, and that cGMP-elevating agents have bone-anabolic effects in mice. Here, we generated mice with an osteoblast-specific (OB) knockout (KO) of type 2 PKG (gene name Prkg2) using a Col1a1(2.3 kb)-Cre driver. Compared to wild type (WT) littermates, 8-week-old male OB Prkg2-KO mice had fewer osteoblasts, reduced bone formation rates, and lower trabecular and cortical bone volumes. Female OB Prkg2-KO littermates showed no bone abnormalities, despite the same degree of PKG2 deficiency in bone. Expression of osteoblast differentiation- and Wnt/β-catenin-related genes was lower in primary osteoblasts and bones of male KO but not female KO mice compared to WT littermates. Osteoclast parameters were unaffected in both sexes. Since PKG2 is part of a mechano-sensitive complex in osteoblast membranes, we examined its role during mechanical loading. Cyclical compression of the tibia increased cortical thickness and induced mechanosensitive and Wnt/β-catenin-related genes to a similar extent in male and female WT mice and female OB Prkg2-KO mice, but loading had a minimal effect in male KO mice. We conclude that PKG2 drives bone acquisition and adaptation to mechanical loading via the Wnt/β-catenin pathway in male mice. The striking sexual dimorphism of OB Prkg2-KO mice suggests that current U.S. Food and Drug Administration-approved cGMP-elevating agents may represent novel effective treatment options for male osteoporosis. © 2022 American Society for Bone and Mineral Research (ASBMR).
Collapse
Affiliation(s)
- Hema Kalyanaraman
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
- These two authors contributed equally to the work
| | - Shyamsundar Pal China
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
- These two authors contributed equally to the work
| | - Justin A. Cabriales
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Jafar Moininazeri
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Darren E. Casteel
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Julian J. Garcia
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
- Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, USA
| | - Van W. Wong
- Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, USA
| | - Albert Chen
- Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, USA
| | - Robert L. Sah
- Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, USA
| | - Gerry R. Boss
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Renate B. Pilz
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| |
Collapse
|
24
|
Michigami T. Paracrine and endocrine functions of osteocytes. Clin Pediatr Endocrinol 2023; 32:1-10. [PMID: 36761497 PMCID: PMC9887291 DOI: 10.1297/cpe.2022-0053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Accepted: 08/30/2022] [Indexed: 11/04/2022] Open
Abstract
Osteocytes are dendritic-shaped cells embedded in the bone matrix and are terminally differentiated from osteoblasts. Inaccessibility due to their location has hindered the understanding of the molecular functions of osteocytes. However, scientific advances in the past few decades have revealed that osteocytes play critical roles in bone and mineral metabolism through their paracrine and endocrine functions. Sclerostin produced by osteocytes regulates bone formation and resorption by inhibiting Wnt/β-catenin signaling in osteoblast-lineage cells. Receptor activator of nuclear factor κ B ligand (RANKL) derived from osteocytes is essential for osteoclastogenesis and osteoclast activation during postnatal life. Osteocytes also secrete fibroblast growth factor 23 (FGF23), an endocrine FGF that regulates phosphate metabolism mainly by increasing phosphate excretion and decreasing 1, 25-dihydroxyvitamin D production in the kidneys. The regulation of FGF23 production in osteocytes is complex and multifactorial, involving many local and systemic regulators. Antibodies against sclerostin, RANKL, and FGF23 have emerged as new strategies for the treatment of metabolic bone diseases. Improved undrstanding of the paracrine and endocrine functions of osteocytes will provide insight into future therapeutic options.
Collapse
Affiliation(s)
- Toshimi Michigami
- Department of Bone and Mineral Research, Research Institute,
Osaka Women’s and Children’s Hospital, Osaka Prefectural Hospital Organization, Osaka,
Japan
| |
Collapse
|
25
|
Wang J, Sun YX, Li J. The role of mechanosensor Piezo1 in bone homeostasis and mechanobiology. Dev Biol 2023; 493:80-88. [PMID: 36368521 DOI: 10.1016/j.ydbio.2022.11.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 10/15/2022] [Accepted: 11/03/2022] [Indexed: 11/09/2022]
Abstract
Bones and articular cartilage are important load-bearing tissues. The fluid flow inside the bone cells and cell interaction with the extracellular matrix serve as the mechanical cues for bones and joints. Piezo1 is an ion channel found on the cell surface of many cell types, including osteocytes and chondrocytes. It is activated in response to mechanical stimulation, which subsequently mediates a variety of signaling pathways in osteoblasts, osteocytes, and chondrocytes. Piezo1 activation in osteoblastic cells positively regulates osteogenesis, while its activation in joints mediates cartilage degradation. This review focuses on the most recent research on Piezo1 in bone development and regeneration.
Collapse
Affiliation(s)
- Jiao Wang
- Department of Anesthesiology, The First Affiliated Hospital of China Medical University, NO.155 Nanjing North Street, Shenyang City, Liaoning Province, 110000, China.
| | - Yong-Xin Sun
- Department of Rehabilitation, The First Affiliated Hospital of China Medical University, NO.155 Nanjing North Street, Shenyang City, Liaoning Province, 110000, China.
| | - Jiliang Li
- Department of Biology, Indiana University Purdue University Indianapolis, 723 West Michigan Street, SL 306, Indianapolis, IN, 46202, USA.
| |
Collapse
|
26
|
Nix Z, Kota D, Ratnayake I, Wang C, Smith S, Wood S. Spectral characterization of cell surface motion for mechanistic investigations of cellular mechanobiology. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2022; 176:3-15. [PMID: 36108781 DOI: 10.1016/j.pbiomolbio.2022.08.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 07/27/2022] [Accepted: 08/05/2022] [Indexed: 06/15/2023]
Abstract
Understanding the specific mechanisms responsible for anabolic and catabolic responses to static or dynamic force are largely poorly understood. Because of this, most research groups studying mechanotransduction due to dynamic forces employ an empirical approach in deciding what frequencies to apply during experiments. While this has been shown to elucidate valuable information regarding how cells respond under controlled provocation, it is often difficult or impossible to determine a true optimal frequency for force application, as many intracellular complexes are involved in receiving, propagating, and responding to a given stimulus. Here we present a novel adaptation of an analytical technique from the fields of civil and mechanical engineering that may open the door to direct measurement of mechanobiological cellular frequencies which could be used to target specific cell signaling pathways leveraging synergy between outside-in and inside-out mechanotransduction approaches. This information could be useful in identifying how specific proteins are involved in the homeostatic balance, or disruption thereof, of cells and tissue, furthering the understanding of the pathogenesis and progression of many diseases across a wide variety of cell types, which may one day lead to the development of novel mechanobiological therapies for clinical use.
Collapse
Affiliation(s)
- Zachary Nix
- Department of Nanoscience & Biomedical Engineering, BioSystems Networks / Translational Research (BioSNTR), South Dakota School of Mines and Technology, USA
| | - Divya Kota
- Department of Nanoscience & Biomedical Engineering, BioSystems Networks / Translational Research (BioSNTR), South Dakota School of Mines and Technology, USA
| | - Ishara Ratnayake
- Department of Nanoscience & Biomedical Engineering, BioSystems Networks / Translational Research (BioSNTR), South Dakota School of Mines and Technology, USA
| | - Congzhou Wang
- Department of Nanoscience & Biomedical Engineering, BioSystems Networks / Translational Research (BioSNTR), South Dakota School of Mines and Technology, USA
| | - Steve Smith
- Department of Nanoscience & Biomedical Engineering, BioSystems Networks / Translational Research (BioSNTR), South Dakota School of Mines and Technology, USA
| | - Scott Wood
- Department of Nanoscience & Biomedical Engineering, BioSystems Networks / Translational Research (BioSNTR), South Dakota School of Mines and Technology, USA.
| |
Collapse
|
27
|
Zeng Y, Riquelme MA, Hua R, Zhang J, Acosta FM, Gu S, Jiang JX. Mechanosensitive piezo1 calcium channel activates connexin 43 hemichannels through PI3K signaling pathway in bone. Cell Biosci 2022; 12:191. [PMID: 36457052 PMCID: PMC9716748 DOI: 10.1186/s13578-022-00929-w] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 11/11/2022] [Indexed: 12/05/2022] Open
Abstract
BACKGROUND Mechanical loading promotes bone formation and osteocytes are a major mechanosensory cell in the bone. Both Piezo1 channels and connexin 43 hemichannels (Cx43 HCs) in osteocytes are important players in mechanotransduction and anabolic function by mechanical loading. However, the mechanism underlying mechanotransduction involving Piezo1 channels and Cx43 HCs in osteocytes and bone remains unknown. RESULTS We showed that, like mechanical loading, Piezo1 specific agonist Yoda1 was able to increase intracellular Ca2+ signaling and activate Cx43 HCs, while Yoda1 antagonist Dooku1 inhibited Ca2+ and Cx43 HC activation induced by both mechanical loading and Yoda1. Moreover, the intracellular Ca2+ signal activated by Yoda1 was reduced by the inhibition of Cx43 HCs and pannexin1 (Panx1) channels, as well as ATP-P2X receptor signaling. Piezo1 and Cx43 HCs were co-localized on the osteocyte cell surface, and Yoda1-activated PI3K-Akt signaling regulated the opening of Cx43 HCs. Furthermore, Cx43 HCs opening by mechanical loading on tibias was ablated by inhibition of Piezo1 activation in vivo. CONCLUSION We demonstrated that upon mechanical stress, increased intracellular Ca2+ activated by Piezo1 regulates the opening of HCs through PI3K-Akt and opened Cx43 HCs, along with Panx1 channels, and ATP-P2X signaling sustain the intracellular Ca2+ signal, leading to bone anabolic function.
Collapse
Affiliation(s)
- Yan Zeng
- Department of Biochemistry and Structural Biology, University of Texas Health Science Center, San Antonio, TX, USA
- The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Manuel A Riquelme
- Department of Biochemistry and Structural Biology, University of Texas Health Science Center, San Antonio, TX, USA
| | - Rui Hua
- Department of Biochemistry and Structural Biology, University of Texas Health Science Center, San Antonio, TX, USA
| | - Jingruo Zhang
- Department of Biochemistry and Structural Biology, University of Texas Health Science Center, San Antonio, TX, USA
| | - Francisca M Acosta
- Department of Biochemistry and Structural Biology, University of Texas Health Science Center, San Antonio, TX, USA
| | - Sumin Gu
- Department of Biochemistry and Structural Biology, University of Texas Health Science Center, San Antonio, TX, USA
| | - Jean X Jiang
- Department of Biochemistry and Structural Biology, University of Texas Health Science Center, San Antonio, TX, USA.
| |
Collapse
|
28
|
Phosphatidylinositol 3-Kinase/Protein Kinase B/Mammalian Target of the Rapamycin Pathway-Related Protein Expression in Lung Squamous Cell Carcinoma and Its Correlation with Lymph Node Metastasis. JOURNAL OF ONCOLOGY 2022; 2022:4537256. [PMID: 36052284 PMCID: PMC9427249 DOI: 10.1155/2022/4537256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 06/29/2022] [Accepted: 07/08/2022] [Indexed: 11/29/2022]
Abstract
The targeted therapy of lung squamous cell carcinoma (LSCC), a pathological type of non-small-cell lung cancer, is relatively lacking by contrast with lung adenocarcinoma. The overexpression or inhibition of intracellular signaling pathways leads to disease. To evaluate genes for a targeted therapy in LSCC, we analyzed PI3K pathway components in LSCC tissues and found elevated PI3K levels in LSCC tissues compared with adjacent counterparts. A comparison of PI3K levels in tissues with and without metastasis revealed that the PI3K pathway activity was greatly increased in metastatic tissues. Our findings suggest that the metastasis of cancer cells in patients with LSCC is closely related to the overexpression of PI3K pathway components in cancer tissues. We performed in vitro cell culture experiments and found that inhibition of PI3K activity decreased proliferation and increased apoptosis in H520 cells, suggesting that PI3K pathway inhibition limits LSCC cell proliferation. We hypothesize that LSCC metastasis is related to the overexpression of PI3K pathway components and inhibiting this pathway may help reduce the risk of lymph node metastasis in LSCC patients.
Collapse
|
29
|
Ono T, Denda R, Tsukahara Y, Nakamura T, Okamoto K, Takayanagi H, Nakashima T. Simultaneous augmentation of muscle and bone by locomomimetism through calcium-PGC-1α signaling. Bone Res 2022; 10:52. [PMID: 35918335 PMCID: PMC9345981 DOI: 10.1038/s41413-022-00225-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 06/01/2022] [Accepted: 06/19/2022] [Indexed: 11/09/2022] Open
Abstract
Impaired locomotion has been extensively studied worldwide because those afflicted with it have a potential risk of becoming bedridden. Physical exercise at times can be an effective remedy for frailty, but exercise therapy cannot be applied in all clinical cases. Medication is safer than exercise, but there are no drugs that reinforce both muscle and bone when administered alone. Multiple medications increase the risk of adverse events; thus, there is a need for individual drugs targeting both tissues. To this end, we established a novel sequential drug screening system and identified an aminoindazole derivative, locamidazole (LAMZ), which promotes both myogenesis and osteoblastogenesis while suppressing osteoclastogenesis. Administration of this drug enhanced locomotor function, with muscle and bone significantly strengthened. Mechanistically, LAMZ induced Mef2c and PGC-1α in a calcium signaling-dependent manner. As this signaling is activated upon physical exercise, LAMZ mimics physical exercise. Thus, LAMZ is a promising therapeutic drug for locomotor diseases, including sarcopenia and osteoporosis.
Collapse
Affiliation(s)
- Takehito Ono
- Department of Cell Signaling, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Yushima 1-5-45, Bunkyo-ku, Tokyo, 113-8549, Japan
| | - Ryosuke Denda
- Department of Cell Signaling, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Yushima 1-5-45, Bunkyo-ku, Tokyo, 113-8549, Japan
- Department of Orthopaedic Surgery, The Jikei University School of Medicine, 3-25-8 Nishi-Shimbashi, Minato-ku, Tokyo, 105-8461, Japan
| | - Yuta Tsukahara
- Department of Cell Signaling, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Yushima 1-5-45, Bunkyo-ku, Tokyo, 113-8549, Japan
- School of Dentistry, Tokyo Medical and Dental University (TMDU), Yushima 1-5-45, Bunkyo-ku, Tokyo, 113-8549, Japan
| | - Takashi Nakamura
- Department of Biochemistry, Tokyo Dental College, Kandamisakicho 2-9-18, Chiyoda-ku, Tokyo, 101-0061, Japan
| | - Kazuo Okamoto
- Department of Osteoimmunology, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, Hongo 7-3-1, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Hiroshi Takayanagi
- Department of Immunology, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, Hongo 7-3-1, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Tomoki Nakashima
- Department of Cell Signaling, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Yushima 1-5-45, Bunkyo-ku, Tokyo, 113-8549, Japan.
| |
Collapse
|
30
|
Xu H, Guan J, Jin Z, Yin C, Wu S, Sun W, Zhang H, Yan B. Mechanical force modulates macrophage proliferation via Piezo1-AKT-Cyclin D1 axis. FASEB J 2022; 36:e22423. [PMID: 35775626 DOI: 10.1096/fj.202200314r] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 05/21/2022] [Accepted: 06/08/2022] [Indexed: 12/23/2022]
Abstract
Orthodontic tooth movement (OTM) is induced by biomechanical stimuli and facilitated by periodontal tissue remodeling, where multiple immune cells participate in this progression. It has been demonstrated that macrophage is essential for mechanical force-induced tissue remodeling. In this study, we first found that mechanical force significantly induced macrophage proliferation in human periodontal samples and murine OTM models. Yet, how macrophages perceive mechanical stimuli and thereby modulate their biological behaviors remain elusive. To illustrate the mechanisms of mechanical force-induced macrophage proliferation, we subsequently identified Piezo1, a novel mechanosensory ion channel, to modulate macrophage response subjected to mechanical stimuli. Mechanical force upregulates Piezo1 expression in periodontal tissues and cultured bone-marrow-derived macrophages (BMDMs). Remarkably, suppressing Piezo1 with GsMTx4 retarded OTM through reduced macrophage proliferation. Moreover, knockdown of Piezo1 effectively inhibited mechanical force-induced BMDMs proliferation. RNA sequencing was further performed to dissect the underlying mechanisms of Piezo1-mediated mechanotransduction utilizing mechanical stretch system. We revealed that Piezo1-activated AKT/GSK3β signaling was closely associated with macrophage proliferation upon mechanical stimuli. Importantly, Cyclin D1 (Ccnd1) was authenticated as a critical downstream factor of Piezo1 that facilitated proliferation by enhancing Rb phosphorylation. We generated genetically modified mice in which Ccnd1 could be deleted in macrophages in an inducible manner. Conditional ablation of Ccnd1 inhibited periodontal macrophage proliferation and therefore delayed OTM. Overall, our findings highlight that proliferation driven by mechanical force is a key process by which macrophages infiltrate in periodontal tissue during OTM, where Piezo1-AKT-Ccnd1 axis plays a pivotal role.
Collapse
Affiliation(s)
- Hao Xu
- Department of Orthodontics, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, China.,Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China.,Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing, China
| | - Jiani Guan
- Department of Orthodontics, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, China.,Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China.,Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing, China
| | - Zhichun Jin
- Department of Orthodontics, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, China.,Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China.,Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing, China
| | - Cheng Yin
- Department of Orthodontics, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, China.,Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China.,Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing, China
| | - Shengnan Wu
- Department of Orthodontics, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, China.,Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China.,Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing, China
| | - Wen Sun
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China
| | - Hanwen Zhang
- School of Basic Medical Sciences, Nanjing Medical University, Nanjing, China.,Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, China
| | - Bin Yan
- Department of Orthodontics, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, China.,Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China.,Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing, China
| |
Collapse
|
31
|
Li X, Hu J, Zhao X, Li J, Chen Y. Piezo channels in the urinary system. Exp Mol Med 2022; 54:697-710. [PMID: 35701561 PMCID: PMC9256749 DOI: 10.1038/s12276-022-00777-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 01/25/2022] [Accepted: 02/16/2022] [Indexed: 12/24/2022] Open
Abstract
The Piezo channel family, including Piezo1 and Piezo2, includes essential mechanosensitive transduction molecules in mammals. Functioning in the conversion of mechanical signals to biological signals to regulate a plethora of physiological processes, Piezo channels, which have a unique homotrimeric three-blade propeller-shaped structure, utilize a cap-motion and plug-and-latch mechanism to gate their ion-conducting pathways. Piezo channels have a wide range of biological roles in various human systems, both in vitro and in vivo. Currently, there is a lack of comprehensive understanding of their antagonists and agonists, and therefore further investigation is needed. Remarkably, increasingly compelling evidence demonstrates that Piezo channel function in the urinary system is important. This review article systematically summarizes the existing evidence of the importance of Piezo channels, including protein structure, mechanogating mechanisms, and pharmacological characteristics, with a particular focus on their physiological and pathophysiological roles in the urinary system. Collectively, this review aims to provide a direction for future clinical applications in urinary system diseases.
Collapse
Affiliation(s)
- Xu Li
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Junwei Hu
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Xuedan Zhao
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Juanjuan Li
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Yuelai Chen
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China.
| |
Collapse
|
32
|
Jia Y, Yao Y, Zhuo L, Chen X, Yan C, Ji Y, Tao J, Zhu Y. Aerobic Physical Exercise as a Non-medical Intervention for Brain Dysfunction: State of the Art and Beyond. Front Neurol 2022; 13:862078. [PMID: 35645958 PMCID: PMC9136296 DOI: 10.3389/fneur.2022.862078] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Accepted: 03/28/2022] [Indexed: 12/03/2022] Open
Abstract
Brain disorders, including stroke, Alzheimer's disease, depression, and chronic pain, are difficult to effectively treat. These major brain disorders have high incidence and mortality rates in the general population, and seriously affect not only the patient's quality of life, but also increases the burden of social medical care. Aerobic physical exercise is considered an effective adjuvant therapy for preventing and treating major brain disorders. Although the underlying regulatory mechanisms are still unknown, systemic processes may be involved. Here, this review aimed to reveal that aerobic physical exercise improved depression and several brain functions, including cognitive functions, and provided chronic pain relief. We concluded that aerobic physical exercise helps to maintain the regulatory mechanisms of brain homeostasis through anti-inflammatory mechanisms and enhanced synaptic plasticity and inhibition of hippocampal atrophy and neuronal apoptosis. In addition, we also discussed the cross-system mechanisms of aerobic exercise in regulating imbalances in brain function, such as the “bone-brain axis.” Furthermore, our findings provide a scientific basis for the clinical application of aerobic physical exercise in the fight against brain disorders.
Collapse
Affiliation(s)
- Yuxiang Jia
- School of Medicine and School of Life Sciences, Shanghai University, Shanghai, China
| | - Yu Yao
- School of Medicine and School of Life Sciences, Shanghai University, Shanghai, China
| | - Limin Zhuo
- School of Medicine and School of Life Sciences, Shanghai University, Shanghai, China
| | - Xingxing Chen
- Department of Neurology and Central Laboratory, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Cuina Yan
- Department of Neurology and Central Laboratory, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yonghua Ji
- School of Medicine and School of Life Sciences, Shanghai University, Shanghai, China
- *Correspondence: Yonghua Ji
| | - Jie Tao
- Department of Neurology and Central Laboratory, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Jie Tao
| | - Yudan Zhu
- Department of Neurology and Central Laboratory, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Yudan Zhu
| |
Collapse
|
33
|
Iwamoto R, Koide M, Udagawa N, Kobayashi Y. Positive and Negative Regulators of Sclerostin Expression. Int J Mol Sci 2022; 23:ijms23094895. [PMID: 35563281 PMCID: PMC9102037 DOI: 10.3390/ijms23094895] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 04/27/2022] [Accepted: 04/27/2022] [Indexed: 02/04/2023] Open
Abstract
Sclerostin is secreted from osteocytes, binds to the Wnt co-receptor Lrp5/6, and affects the interaction between Wnt ligands and Lrp5/6, which inhibits Wnt/β-catenin signals and suppresses bone formation. Sclerostin plays an important role in the preservation of bone mass by functioning as a negative regulator of bone formation. A sclerostin deficiency causes sclerosteosis, which is characterized by an excess bone mass with enhanced bone formation in humans and mice. The expression of sclerostin is positively and negatively regulated by many factors, which also govern bone metabolism. Positive and negative regulators of sclerostin expression and their effects are introduced and discussed herein based on recent and previous findings, including our research.
Collapse
Affiliation(s)
- Rina Iwamoto
- Division of Hard Tissue Research, Institute for Oral Science, Matsumoto Dental University, 1780 Gobara Hiro-oka, Shiojiri 399-0781, Nagano, Japan; (R.I.); (M.K.)
| | - Masanori Koide
- Division of Hard Tissue Research, Institute for Oral Science, Matsumoto Dental University, 1780 Gobara Hiro-oka, Shiojiri 399-0781, Nagano, Japan; (R.I.); (M.K.)
| | - Nobuyuki Udagawa
- Department of Biochemistry, Matsumoto Dental University, 1780 Gobara Hiro-oka, Shiojiri 399-0781, Nagano, Japan;
| | - Yasuhiro Kobayashi
- Division of Hard Tissue Research, Institute for Oral Science, Matsumoto Dental University, 1780 Gobara Hiro-oka, Shiojiri 399-0781, Nagano, Japan; (R.I.); (M.K.)
- Correspondence: ; Tel.: +81-263-51-2238
| |
Collapse
|
34
|
Sclerostin: From Molecule to Clinical Biomarker. Int J Mol Sci 2022; 23:ijms23094751. [PMID: 35563144 PMCID: PMC9104784 DOI: 10.3390/ijms23094751] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 04/12/2022] [Accepted: 04/22/2022] [Indexed: 12/23/2022] Open
Abstract
Sclerostin, a glycoprotein encoded by the SOST gene, is mainly produced by mature osteocytes and is a critical regulator of bone formation through its inhibitory effect on Wnt signaling. Osteocytes are differentiated osteoblasts that form a vast and highly complex communication network and orchestrate osteogenesis in response to both mechanical and hormonal cues. The three most commonly described pathways of SOST gene regulation are mechanotransduction, Wnt/β-catenin, and steroid signaling. Downregulation of SOST and thereby upregulation of local Wnt signaling is required for the osteogenic response to mechanical loading. This review covers recent findings concerning the identification of SOST, in vitro regulation of SOST gene expression, structural and functional properties of sclerostin, pathophysiology, biological variability, and recent assay developments for measuring circulating sclerostin. The three-dimensional structure of human sclerostin was generated with the AlphaFold Protein Structure Database applying a novel deep learning algorithm based on the amino acid sequence. The functional properties of the 3-loop conformation within the tertiary structure of sclerostin and molecular interaction with low-density lipoprotein receptor-related protein 6 (LRP6) are also reviewed. Second-generation immunoassays for intact/biointact sclerostin have recently been developed, which might overcome some of the reported methodological obstacles. Sclerostin assay standardization would be a long-term objective to overcome some of the problems with assay discrepancies. Besides the use of age- and sex-specific reference intervals for sclerostin, it is also pivotal to use assay-specific reference intervals since available immunoassays vary widely in their methodological characteristics.
Collapse
|
35
|
Maeda K, Yoshida K, Nishizawa T, Otani K, Yamashita Y, Okabe H, Hadano Y, Kayama T, Kurosaka D, Saito M. Inflammation and Bone Metabolism in Rheumatoid Arthritis: Molecular Mechanisms of Joint Destruction and Pharmacological Treatments. Int J Mol Sci 2022; 23:2871. [PMID: 35270012 PMCID: PMC8911191 DOI: 10.3390/ijms23052871] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 02/28/2022] [Accepted: 03/02/2022] [Indexed: 12/31/2022] Open
Abstract
Rheumatoid arthritis (RA) is an inflammatory disease characterized by a variety of symptoms and pathologies often presenting with polyarthritis. The primary symptom in the initial stage is joint swelling due to synovitis. With disease progression, cartilage and bone are affected to cause joint deformities. Advanced osteoarticular destruction and deformation can cause irreversible physical disabilities. Physical disabilities not only deteriorate patients' quality of life but also have substantial medical economic effects on society. Therefore, prevention of the progression of osteoarticular destruction and deformation is an important task. Recent studies have progressively improved our understanding of the molecular mechanism by which synovitis caused by immune disorders results in activation of osteoclasts; activated osteoclasts in turn cause bone destruction and para-articular osteoporosis. In this paper, we review the mechanisms of bone metabolism under physiological and RA conditions, and we describe the effects of therapeutic intervention against RA on bone.
Collapse
Affiliation(s)
- Kazuhiro Maeda
- Department of Orthopaedic Surgery, The Jikei University School of Medicine, 3-25-8 Nishi-Shimbashi, Minato-ku, Tokyo 105-8461, Japan; (T.N.); (Y.Y.); (H.O.); (Y.H.); (T.K.); (M.S.)
| | - Ken Yoshida
- Division of Rheumatology, Department of Internal Medicine, The Jikei University School of Medicine, 3-25-8 Nishi-Shimbashi, Minato-ku, Tokyo 105-8461, Japan; (K.Y.); (K.O.); (D.K.)
| | - Tetsuro Nishizawa
- Department of Orthopaedic Surgery, The Jikei University School of Medicine, 3-25-8 Nishi-Shimbashi, Minato-ku, Tokyo 105-8461, Japan; (T.N.); (Y.Y.); (H.O.); (Y.H.); (T.K.); (M.S.)
| | - Kazuhiro Otani
- Division of Rheumatology, Department of Internal Medicine, The Jikei University School of Medicine, 3-25-8 Nishi-Shimbashi, Minato-ku, Tokyo 105-8461, Japan; (K.Y.); (K.O.); (D.K.)
| | - Yu Yamashita
- Department of Orthopaedic Surgery, The Jikei University School of Medicine, 3-25-8 Nishi-Shimbashi, Minato-ku, Tokyo 105-8461, Japan; (T.N.); (Y.Y.); (H.O.); (Y.H.); (T.K.); (M.S.)
| | - Hinako Okabe
- Department of Orthopaedic Surgery, The Jikei University School of Medicine, 3-25-8 Nishi-Shimbashi, Minato-ku, Tokyo 105-8461, Japan; (T.N.); (Y.Y.); (H.O.); (Y.H.); (T.K.); (M.S.)
| | - Yuka Hadano
- Department of Orthopaedic Surgery, The Jikei University School of Medicine, 3-25-8 Nishi-Shimbashi, Minato-ku, Tokyo 105-8461, Japan; (T.N.); (Y.Y.); (H.O.); (Y.H.); (T.K.); (M.S.)
| | - Tomohiro Kayama
- Department of Orthopaedic Surgery, The Jikei University School of Medicine, 3-25-8 Nishi-Shimbashi, Minato-ku, Tokyo 105-8461, Japan; (T.N.); (Y.Y.); (H.O.); (Y.H.); (T.K.); (M.S.)
| | - Daitaro Kurosaka
- Division of Rheumatology, Department of Internal Medicine, The Jikei University School of Medicine, 3-25-8 Nishi-Shimbashi, Minato-ku, Tokyo 105-8461, Japan; (K.Y.); (K.O.); (D.K.)
| | - Mitsuru Saito
- Department of Orthopaedic Surgery, The Jikei University School of Medicine, 3-25-8 Nishi-Shimbashi, Minato-ku, Tokyo 105-8461, Japan; (T.N.); (Y.Y.); (H.O.); (Y.H.); (T.K.); (M.S.)
| |
Collapse
|
36
|
Kloen P, Loots G, Hamdy R, Smit T. Bridging the gap: compressing non-unions for proper cellular signaling. Med Hypotheses 2022. [DOI: 10.1016/j.mehy.2022.110794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
37
|
Abstract
Osteocytes are dendritic cells in the mineralized bone matrix that descend from osteoblasts. They play critical roles in controlling bone mass through the production of sclerostin, an inhibitor of bone formation, and receptor activator of nuclear factor κ B ligand, an inducer of osteoblastic bone resorption. Osteocytes also govern phosphate homeostasis through the production of fibroblast growth factor 23 (FGF23), which lowers serum phosphate levels by increasing renal phosphate excretion and reducing the synthesis of 1,25-dihydroxyvitamin D (1,25(OH)2D), an active metabolite of vitamin D. The production of FGF23 in osteocytes is regulated by various local and systemic factors. Phosphate-regulating gene homologous to endopeptidase on X chromosome (PHEX), dentin matrix protein 1 (DMP1), and family with sequence similarity 20, member C function as local negative regulators of FGF23 production in osteocytes, and their inactivation causes the overproduction of FGF23 and hypophosphatemia. Sclerostin has been suggested to regulate the production of FGF23, which may link the two functions of osteocytes, namely, the control of bone mass and regulation of phosphate homeostasis. Systemic regulators of FGF23 production include 1,25(OH)2D, phosphate, parathyroid hormone, insulin, iron, and inflammation. Therefore, the regulation of FGF23 in osteocytes is complex and multifactorial. Recent mouse studies have suggested that decreases in serum phosphate levels from youth to adulthood are caused by growth-related increases in FGF23 production by osteocytes, which are associated with the down-regulation of Phex and Dmp1.
Collapse
|
38
|
Yamazaki M, Michigami T. Osteocytes and the pathogenesis of hypophosphatemic rickets. Front Endocrinol (Lausanne) 2022; 13:1005189. [PMID: 36246908 PMCID: PMC9556901 DOI: 10.3389/fendo.2022.1005189] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Accepted: 09/15/2022] [Indexed: 11/13/2022] Open
Abstract
Since phosphorus is a component of hydroxyapatite, its prolonged deprivation affects bone mineralization. Fibroblast growth factor 23 (FGF23) is essential for maintaining phosphate homeostasis and is mainly produced by osteocytes. FGF23 increases the excretion of inorganic phosphate (Pi) and decreases the production of 1,25-dihydroxyvitamin D in the kidneys. Osteocytes are cells of osteoblastic lineage that have undergone terminal differentiation and become embedded in mineralized bone matrix. Osteocytes express FGF23 and other multiple genes responsible for hereditary hypophosphatemic rickets, which include phosphate-regulating gene homologous to endopeptidase on X chromosome (PHEX), dentin matrix protein 1 (DMP1), and family with sequence similarity 20, member C (FAM20C). Since inactivating mutations in PHEX, DMP1, and FAM20C boost the production of FGF23, these molecules might be considered as local negative regulators of FGF23. Mouse studies have suggested that enhanced FGF receptor (FGFR) signaling is involved in the overproduction of FGF23 in PHEX-deficient X-linked hypophosphatemic rickets (XLH) and DMP1-deficient autosomal recessive hypophosphatemic rickets type 1. Since FGFR is involved in the transduction of signals evoked by extracellular Pi, Pi sensing in osteocytes may be abnormal in these diseases. Serum levels of sclerostin, an inhibitor Wnt/β-catenin signaling secreted by osteocytes, are increased in XLH patients, and mouse studies have suggested the potential of inhibiting sclerostin as a new therapeutic option for the disease. The elucidation of complex abnormalities in the osteocytes of FGF23-related hypophosphatemic diseases will provide a more detailed understanding of their pathogenesis and more effective treatments.
Collapse
|
39
|
Kameo Y, Ozasa M, Adachi T. Computational framework for analyzing flow-induced strain on osteocyte as modulated by microenvironment. J Mech Behav Biomed Mater 2021; 126:105027. [PMID: 34920322 DOI: 10.1016/j.jmbbm.2021.105027] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 11/19/2021] [Accepted: 12/02/2021] [Indexed: 12/26/2022]
Abstract
Osteocytes buried in bone matrix are major mechanosensory cells that regulate bone remodeling in response to interstitial fluid flow in a lacuno-canalicular porosity. To gain an understanding of the mechanism of osteocyte mechanosensing, it is important to be able to evaluate the local strain on the osteocyte process membrane induced by the interstitial fluid flow. The microenvironment of the osteocytes, including the pericellular matrix (PCM) and canalicular ultrastructure, is a key modulator of the flow-induced strain on the osteocyte process membrane because it produces heterogeneous flow patterns in the pericellular space. To investigate the effect of changes in the microenvironment of osteocytes on the flow-induced strain, we developed a novel computational framework for analyzing the fluid-structure interaction. Computer simulations based on the proposed framework enabled evaluation of the spatial distribution of flow-induced strain on the osteocyte process membrane according to changes in the PCM density and canalicular curvature. The simulation results reveal that a decrease in PCM density and an increase in canalicular curvature, each of which is associated with aging and bone disease, have the notable effect of enhancing local flow-induced strain on the osteocyte process membrane. We believe that the proposed computational framework is a promising framework for investigating cell-specific mechanical stimuli and that it has the potential to accelerate the mechanobiological study of osteocytes by providing a deeper understanding of their mechanical environment in living bone tissue.
Collapse
Affiliation(s)
- Yoshitaka Kameo
- Department of Biosystems Science, Institute for Frontier Life and Medical Sciences, Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan; Department of Micro Engineering, Graduate School of Engineering, Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan; Department of Mammalian Regulatory Network, Graduate School of Biostudies, Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan.
| | - Masahiro Ozasa
- Department of Micro Engineering, Graduate School of Engineering, Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Taiji Adachi
- Department of Biosystems Science, Institute for Frontier Life and Medical Sciences, Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan; Department of Micro Engineering, Graduate School of Engineering, Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan; Department of Mammalian Regulatory Network, Graduate School of Biostudies, Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| |
Collapse
|
40
|
Lewis KJ. Osteocyte calcium signaling - A potential translator of mechanical load to mechanobiology. Bone 2021; 153:116136. [PMID: 34339908 DOI: 10.1016/j.bone.2021.116136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 06/25/2021] [Accepted: 07/27/2021] [Indexed: 10/20/2022]
Abstract
Osteocytes are embedded dendritic bone cells; by virtue of their position in bone tissue, ability to coordinate bone building osteoblasts and resorbing osteoclasts, and sensitivity to tissue level mechanical loading, they serve as the resident bone mechanosensor. The mechanisms osteocytes use to change mechanical loading into biological signals that drive tissue level changes has been well studied over the last 30 years, however the ways loading parameters are encoded at the cellular level are still not fully understood. Calcium signaling is a first messenger signal exhibited by osteocytes in response to mechanical forces. A body of work interrogating the mechanisms of osteocyte calcium signaling exists and is presently expanding, presenting the opportunity to better understand the relationship between calcium signaling characteristics and tuned osteocyte responses to tissue level strain features (e.g. magnitude, duration, frequency). This review covers the history of osteocyte load induced calcium signaling and highlights potential cellular mechanisms used by osteocytes to turn details about loading parameters into biological events.
Collapse
Affiliation(s)
- Karl J Lewis
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, United States of America.
| |
Collapse
|
41
|
The mechanosensory and mechanotransductive processes mediated by ion channels and the impact on bone metabolism: A systematic review. Arch Biochem Biophys 2021; 711:109020. [PMID: 34461086 DOI: 10.1016/j.abb.2021.109020] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 08/25/2021] [Accepted: 08/26/2021] [Indexed: 02/06/2023]
Abstract
Mechanical environments were associated with alterations in bone metabolism. Ion channels present on bone cells are indispensable for bone metabolism and can be directly or indirectly activated by mechanical stimulation. This review aimed to discuss the literature reporting the mechanical regulatory effects of ion channels on bone cells and bone tissue. An electronic search was conducted in PubMed, Embase and Web of Science. Studies about mechanically induced alteration of bone cells and bone tissue by ion channels were included. Ion channels including TRP family channels, Ca2+ release-activated Ca2+ channels (CRACs), Piezo1/2 channels, purinergic receptors, NMDA receptors, voltage-sensitive calcium channels (VSCCs), TREK2 potassium channels, calcium- and voltage-dependent big conductance potassium (BKCa) channels, small conductance, calcium-activated potassium (SKCa) channels and epithelial sodium channels (ENaCs) present on bone cells and bone tissue participate in the mechanical regulation of bone development in addition to contributing to direct or indirect mechanotransduction such as altered membrane potential and ionic flux. Physiological (beneficial) mechanical stimulation could induce the anabolism of bone cells and bone tissue through ion channels, but abnormal (harmful) mechanical stimulation could also induce the catabolism of bone cells and bone tissue through ion channels. Functional expression of ion channels is vital for the mechanotransduction of bone cells. Mechanical activation (opening) of ion channels triggers ion influx and induces the activation of intracellular modulators that can influence bone metabolism. Therefore, mechanosensitive ion channels provide new insights into therapeutic targets for the treatment of bone-related diseases such as osteopenia and aseptic implant loosening.
Collapse
|
42
|
Qin L, He T, Chen S, Yang D, Yi W, Cao H, Xiao G. Roles of mechanosensitive channel Piezo1/2 proteins in skeleton and other tissues. Bone Res 2021; 9:44. [PMID: 34667178 PMCID: PMC8526690 DOI: 10.1038/s41413-021-00168-8] [Citation(s) in RCA: 86] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 06/16/2021] [Accepted: 07/08/2021] [Indexed: 12/12/2022] Open
Abstract
Mechanotransduction is a fundamental ability that allows living organisms to receive and respond to physical signals from both the external and internal environments. The mechanotransduction process requires a range of special proteins termed mechanotransducers to convert mechanical forces into biochemical signals in cells. The Piezo proteins are mechanically activated nonselective cation channels and the largest plasma membrane ion channels reported thus far. The regulation of two family members, Piezo1 and Piezo2, has been reported to have essential functions in mechanosensation and transduction in different organs and tissues. Recently, the predominant contributions of the Piezo family were reported to occur in the skeletal system, especially in bone development and mechano-stimulated bone homeostasis. Here we review current studies focused on the tissue-specific functions of Piezo1 and Piezo2 in various backgrounds with special highlights on their importance in regulating skeletal cell mechanotransduction. In this review, we emphasize the diverse functions of Piezo1 and Piezo2 and related signaling pathways in osteoblast lineage cells and chondrocytes. We also summarize our current understanding of Piezo channel structures and the key findings about PIEZO gene mutations in human diseases.
Collapse
Affiliation(s)
- Lei Qin
- Department of Orthopedics, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen, Guangdong, China
- Department of Biochemistry, School of Medicine, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Tailin He
- Department of Biochemistry, School of Medicine, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Sheng Chen
- Department of Biochemistry, School of Medicine, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, Southern University of Science and Technology, Shenzhen, Guangdong, China
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Dazhi Yang
- Department of Orthopedics, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen, Guangdong, China
| | - Weihong Yi
- Department of Orthopedics, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen, Guangdong, China.
| | - Huiling Cao
- Department of Biochemistry, School of Medicine, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, Southern University of Science and Technology, Shenzhen, Guangdong, China.
| | - Guozhi Xiao
- Department of Biochemistry, School of Medicine, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, Southern University of Science and Technology, Shenzhen, Guangdong, China.
| |
Collapse
|
43
|
Poole K. The Diverse Physiological Functions of Mechanically Activated Ion Channels in Mammals. Annu Rev Physiol 2021; 84:307-329. [PMID: 34637325 DOI: 10.1146/annurev-physiol-060721-100935] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Many aspects of mammalian physiology are mechanically regulated. One set of molecules that can mediate mechanotransduction are the mechanically activated ion channels. These ionotropic force sensors are directly activated by mechanical inputs, resulting in ionic flux across the plasma membrane. While there has been much research focus on the role of mechanically activated ion channels in touch sensation and hearing, recent data have highlighted the broad expression pattern of these molecules in mammalian cells. Disruption of mechanically activated channels has been shown to impact (a) the development of mechanoresponsive structures, (b) acute mechanical sensing, and (c) mechanically driven homeostatic maintenance in multiple tissue types. The diversity of processes impacted by these molecules highlights the importance of mechanically activated ion channels in mammalian physiology. Expected final online publication date for the Annual Review of Physiology, Volume 84 is February 2022. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Kate Poole
- EMBL Australia Node in Single Molecule Science, School of Medical Sciences, Faculty of Medicine and Health, University of New South Wales, Sydney, Australia; .,Cellular and Systems Physiology, School of Medical Sciences, Faculty of Medicine and Health, University of New South Wales, Sydney, Australia
| |
Collapse
|
44
|
Role of K + and Ca 2+-Permeable Channels in Osteoblast Functions. Int J Mol Sci 2021; 22:ijms221910459. [PMID: 34638799 PMCID: PMC8509041 DOI: 10.3390/ijms221910459] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 09/23/2021] [Accepted: 09/24/2021] [Indexed: 12/20/2022] Open
Abstract
Bone-forming cells or osteoblasts play an important role in bone modeling and remodeling processes. Osteoblast differentiation or osteoblastogenesis is orchestrated by multiple intracellular signaling pathways (e.g., bone morphogenetic proteins (BMP) and Wnt signaling pathways) and is modulated by the extracellular environment (e.g., parathyroid hormone (PTH), vitamin D, transforming growth factor β (TGF-β), and integrins). The regulation of bone homeostasis depends on the proper differentiation and function of osteoblast lineage cells from osteogenic precursors to osteocytes. Intracellular Ca2+ signaling relies on the control of numerous processes in osteoblast lineage cells, including cell growth, differentiation, migration, and gene expression. In addition, hyperpolarization via the activation of K+ channels indirectly promotes Ca2+ signaling in osteoblast lineage cells. An improved understanding of the fundamental physiological and pathophysiological processes in bone homeostasis requires detailed investigations of osteoblast lineage cells. This review summarizes the current knowledge on the functional impacts of K+ channels and Ca2+-permeable channels, which critically regulate Ca2+ signaling in osteoblast lineage cells to maintain bone homeostasis.
Collapse
|
45
|
Yokoyama Y, Kameo Y, Kamioka H, Adachi T. High-resolution image-based simulation reveals membrane strain concentration on osteocyte processes caused by tethering elements. Biomech Model Mechanobiol 2021; 20:2353-2360. [PMID: 34471950 PMCID: PMC8595188 DOI: 10.1007/s10237-021-01511-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Accepted: 08/13/2021] [Indexed: 11/08/2022]
Abstract
Osteocytes are vital for regulating bone remodeling by sensing the flow-induced mechanical stimuli applied to their cell processes. In this mechanosensing mechanism, tethering elements (TEs) connecting the osteocyte process with the canalicular wall potentially amplify the strain on the osteocyte processes. The ultrastructure of the osteocyte processes and canaliculi can be visualized at a nanometer scale using high-resolution imaging via ultra-high voltage electron microscopy (UHVEM). Moreover, the irregular shapes of the osteocyte processes and the canaliculi, including the TEs in the canalicular space, should considerably influence the mechanical stimuli applied to the osteocytes. This study aims to characterize the roles of the ultrastructure of osteocyte processes and canaliculi in the mechanism of osteocyte mechanosensing. Thus, we constructed a high-resolution image-based model of an osteocyte process and a canaliculus using UHVEM tomography and investigated the distribution and magnitude of flow-induced local strain on the osteocyte process by performing fluid–structure interaction simulation. The analysis results reveal that local strain concentration in the osteocyte process was induced by a small number of TEs with high tension, which were inclined depending on the irregular shapes of osteocyte processes and canaliculi. Therefore, this study could provide meaningful insights into the effect of ultrastructure of osteocyte processes and canaliculi on the osteocyte mechanosensing mechanism.
Collapse
Affiliation(s)
- Yuka Yokoyama
- Department of Micro Engineering, Graduate School of Engineering, Kyoto University, 53 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Yoshitaka Kameo
- Department of Micro Engineering, Graduate School of Engineering, Kyoto University, 53 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto, 606-8507, Japan.,Department of Biosystems Science, Institute for Frontier Life and Medical Sciences, Kyoto University, 53 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto, 606-8507, Japan.,Department of Mammalian Regulatory Network, Graduate School of Biostudies, Kyoto University, 53 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Hiroshi Kamioka
- Department of Orthodontics, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, 2-5-1 Shikata-Cho, Kita-ku, Okayama, 700-8525, Japan
| | - Taiji Adachi
- Department of Micro Engineering, Graduate School of Engineering, Kyoto University, 53 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto, 606-8507, Japan. .,Department of Biosystems Science, Institute for Frontier Life and Medical Sciences, Kyoto University, 53 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto, 606-8507, Japan. .,Department of Mammalian Regulatory Network, Graduate School of Biostudies, Kyoto University, 53 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto, 606-8507, Japan.
| |
Collapse
|
46
|
Hagan ML, Balayan V, McGee-Lawrence ME. Plasma membrane disruption (PMD) formation and repair in mechanosensitive tissues. Bone 2021; 149:115970. [PMID: 33892174 PMCID: PMC8217198 DOI: 10.1016/j.bone.2021.115970] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 03/26/2021] [Accepted: 04/17/2021] [Indexed: 01/04/2023]
Abstract
Mammalian cells employ an array of biological mechanisms to detect and respond to mechanical loading in their environment. One such mechanism is the formation of plasma membrane disruptions (PMD), which foster a molecular flux across cell membranes that promotes tissue adaptation. Repair of PMD through an orchestrated activity of molecular machinery is critical for cell survival, and the rate of PMD repair can affect downstream cellular signaling. PMD have been observed to influence the mechanical behavior of skin, alveolar, and gut epithelial cells, aortic endothelial cells, corneal keratocytes and epithelial cells, cardiac and skeletal muscle myocytes, neurons, and most recently, bone cells including osteoblasts, periodontal ligament cells, and osteocytes. PMD are therefore positioned to affect the physiological behavior of a wide range of vertebrate organ systems including skeletal and cardiac muscle, skin, eyes, the gastrointestinal tract, the vasculature, the respiratory system, and the skeleton. The purpose of this review is to describe the processes of PMD formation and repair across these mechanosensitive tissues, with a particular emphasis on comparing and contrasting repair mechanisms and downstream signaling to better understand the role of PMD in skeletal mechanobiology. The implications of PMD-related mechanisms for disease and potential therapeutic applications are also explored.
Collapse
Affiliation(s)
- Mackenzie L Hagan
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, 1460 Laney Walker Blvd., CB1101, Augusta, GA, USA
| | - Vanshika Balayan
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, 1460 Laney Walker Blvd., CB1101, Augusta, GA, USA
| | - Meghan E McGee-Lawrence
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, 1460 Laney Walker Blvd., CB1101, Augusta, GA, USA; Department of Orthopaedic Surgery, Augusta University, Augusta, GA, USA.
| |
Collapse
|
47
|
Shimohira T, Niimi H, Ohsugi Y, Tsuchiya Y, Morita K, Yoshida S, Hatasa M, Shiba T, Kadokura H, Yokose S, Katagiri S, Iwata T, Aoki A. Low-Level Erbium-Doped Yttrium Aluminum Garnet Laser Irradiation Induced Alteration of Gene Expression in Osteogenic Cells from Rat Calvariae. PHOTOBIOMODULATION PHOTOMEDICINE AND LASER SURGERY 2021; 39:566-577. [PMID: 34339325 DOI: 10.1089/photob.2020.4958] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Objective: The aim of this study was to investigate the effect of low-level erbium-doped yttrium aluminum garnet (Er:YAG) laser irradiation on gene expression in osteogenic cells from rat calvariae. Background: Previous studies showed beneficial effects of laser irradiation on bone-related cells. However, few studies have examined the gene expression alteration by laser irradiation on osteogenic cells in a calcified condition. Materials and methods: Osteogenic cells were prepared by culturing rat calvarial osteoblast-like cells in osteoinductive medium for 21 days. The cells at the bottom of the culture dish were irradiated with Er:YAG laser (wavelength: 2.94 μm, energy density: 3.1 and 8.2 J/cm2) positioned at distance of 25 cm. Lactate dehydrogenase (LDH) assay of the irradiated cells was performed. After screening for genes related to bone formation, mechanotransduction, and thermal effect by quantitative polymerase chain reaction (qPCR), gene expression at 3 h after 3.1 J/cm2 irradiation was comprehensively analyzed using microarray. Results: No dramatical increase in surface temperature and LDH activities after laser irradiation were observed. Sost expression was significantly reduced at 3 h after 3.1 J/cm2 irradiation. Bcar1 and Hspa1a expression was significantly increased following 8.2 J/cm2 irradiation. Microarray analysis identified 116 differentially expressed genes. Gene set enrichment analysis showed enrichment of histone H3-K9 methylation and modification gene sets. Conclusions: Er:YAG laser irradiation, especially at 3.1 J/cm2, showed positive effect on the expression of genes related to bone formation in osteogenic cells, without inducing significant cell damage. These findings may represent critical mechanisms of early bone formation after Er:YAG laser irradiation.
Collapse
Affiliation(s)
- Tsuyoshi Shimohira
- Department of Periodontology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Hiromi Niimi
- Department of Periodontology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Yujin Ohsugi
- Department of Periodontology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Yosuke Tsuchiya
- Department of Periodontology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Kazuki Morita
- Department of Periodontology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Sumiko Yoshida
- Department of Periodontology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Masahiro Hatasa
- Department of Periodontology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Takahiko Shiba
- Department of Periodontology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Hiroshi Kadokura
- Division of Endodontic and Operative Dentistry, Department of Restorative and Biomaterials Sciences, School of Dentistry, Meikai University, Saitama, Japan
| | - Satoshi Yokose
- Division of Endodontic and Operative Dentistry, Department of Restorative and Biomaterials Sciences, School of Dentistry, Meikai University, Saitama, Japan
| | - Sayaka Katagiri
- Department of Periodontology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Takanori Iwata
- Department of Periodontology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Akira Aoki
- Department of Periodontology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| |
Collapse
|
48
|
Xu X, Liu S, Liu H, Ru K, Jia Y, Wu Z, Liang S, Khan Z, Chen Z, Qian A, Hu L. Piezo Channels: Awesome Mechanosensitive Structures in Cellular Mechanotransduction and Their Role in Bone. Int J Mol Sci 2021; 22:ijms22126429. [PMID: 34208464 PMCID: PMC8234635 DOI: 10.3390/ijms22126429] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 06/10/2021] [Accepted: 06/12/2021] [Indexed: 12/13/2022] Open
Abstract
Piezo channels are mechanosensitive ion channels located in the cell membrane and function as key cellular mechanotransducers for converting mechanical stimuli into electrochemical signals. Emerged as key molecular detectors of mechanical forces, Piezo channels' functions in bone have attracted more and more attention. Here, we summarize the current knowledge of Piezo channels and review the research advances of Piezo channels' function in bone by highlighting Piezo1's role in bone cells, including osteocyte, bone marrow mesenchymal stem cell (BM-MSC), osteoblast, osteoclast, and chondrocyte. Moreover, the role of Piezo channels in bone diseases is summarized.
Collapse
Affiliation(s)
- Xia Xu
- Lab for Bone Metabolism, Key Lab for Space Biosciences and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi’an 710072, China; (X.X.); (S.L.); (H.L.); (K.R.); (Y.J.); (Z.W.); (S.L.); (Z.K.); (Z.C.)
- Xi’an Key Laboratory of Special Medicine and Health Engineering, School of Life Sciences, Northwestern Polytechnical University, Xi’an 710072, China
- Research Center for Special Medicine and Health Systems Engineering, School of Life Sciences, Northwestern Polytechnical University, Xi’an 710072, China
- NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi’an 710072, China
| | - Shuyu Liu
- Lab for Bone Metabolism, Key Lab for Space Biosciences and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi’an 710072, China; (X.X.); (S.L.); (H.L.); (K.R.); (Y.J.); (Z.W.); (S.L.); (Z.K.); (Z.C.)
- Xi’an Key Laboratory of Special Medicine and Health Engineering, School of Life Sciences, Northwestern Polytechnical University, Xi’an 710072, China
- Research Center for Special Medicine and Health Systems Engineering, School of Life Sciences, Northwestern Polytechnical University, Xi’an 710072, China
- NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi’an 710072, China
| | - Hua Liu
- Lab for Bone Metabolism, Key Lab for Space Biosciences and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi’an 710072, China; (X.X.); (S.L.); (H.L.); (K.R.); (Y.J.); (Z.W.); (S.L.); (Z.K.); (Z.C.)
- Xi’an Key Laboratory of Special Medicine and Health Engineering, School of Life Sciences, Northwestern Polytechnical University, Xi’an 710072, China
- Research Center for Special Medicine and Health Systems Engineering, School of Life Sciences, Northwestern Polytechnical University, Xi’an 710072, China
- NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi’an 710072, China
| | - Kang Ru
- Lab for Bone Metabolism, Key Lab for Space Biosciences and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi’an 710072, China; (X.X.); (S.L.); (H.L.); (K.R.); (Y.J.); (Z.W.); (S.L.); (Z.K.); (Z.C.)
- Xi’an Key Laboratory of Special Medicine and Health Engineering, School of Life Sciences, Northwestern Polytechnical University, Xi’an 710072, China
- Research Center for Special Medicine and Health Systems Engineering, School of Life Sciences, Northwestern Polytechnical University, Xi’an 710072, China
- NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi’an 710072, China
| | - Yunxian Jia
- Lab for Bone Metabolism, Key Lab for Space Biosciences and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi’an 710072, China; (X.X.); (S.L.); (H.L.); (K.R.); (Y.J.); (Z.W.); (S.L.); (Z.K.); (Z.C.)
- Xi’an Key Laboratory of Special Medicine and Health Engineering, School of Life Sciences, Northwestern Polytechnical University, Xi’an 710072, China
- Research Center for Special Medicine and Health Systems Engineering, School of Life Sciences, Northwestern Polytechnical University, Xi’an 710072, China
- NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi’an 710072, China
| | - Zixiang Wu
- Lab for Bone Metabolism, Key Lab for Space Biosciences and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi’an 710072, China; (X.X.); (S.L.); (H.L.); (K.R.); (Y.J.); (Z.W.); (S.L.); (Z.K.); (Z.C.)
- Xi’an Key Laboratory of Special Medicine and Health Engineering, School of Life Sciences, Northwestern Polytechnical University, Xi’an 710072, China
- Research Center for Special Medicine and Health Systems Engineering, School of Life Sciences, Northwestern Polytechnical University, Xi’an 710072, China
- NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi’an 710072, China
| | - Shujing Liang
- Lab for Bone Metabolism, Key Lab for Space Biosciences and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi’an 710072, China; (X.X.); (S.L.); (H.L.); (K.R.); (Y.J.); (Z.W.); (S.L.); (Z.K.); (Z.C.)
- Xi’an Key Laboratory of Special Medicine and Health Engineering, School of Life Sciences, Northwestern Polytechnical University, Xi’an 710072, China
- Research Center for Special Medicine and Health Systems Engineering, School of Life Sciences, Northwestern Polytechnical University, Xi’an 710072, China
- NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi’an 710072, China
| | - Zarnaz Khan
- Lab for Bone Metabolism, Key Lab for Space Biosciences and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi’an 710072, China; (X.X.); (S.L.); (H.L.); (K.R.); (Y.J.); (Z.W.); (S.L.); (Z.K.); (Z.C.)
- Xi’an Key Laboratory of Special Medicine and Health Engineering, School of Life Sciences, Northwestern Polytechnical University, Xi’an 710072, China
- Research Center for Special Medicine and Health Systems Engineering, School of Life Sciences, Northwestern Polytechnical University, Xi’an 710072, China
- NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi’an 710072, China
| | - Zhihao Chen
- Lab for Bone Metabolism, Key Lab for Space Biosciences and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi’an 710072, China; (X.X.); (S.L.); (H.L.); (K.R.); (Y.J.); (Z.W.); (S.L.); (Z.K.); (Z.C.)
- Xi’an Key Laboratory of Special Medicine and Health Engineering, School of Life Sciences, Northwestern Polytechnical University, Xi’an 710072, China
- Research Center for Special Medicine and Health Systems Engineering, School of Life Sciences, Northwestern Polytechnical University, Xi’an 710072, China
- NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi’an 710072, China
| | - Airong Qian
- Lab for Bone Metabolism, Key Lab for Space Biosciences and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi’an 710072, China; (X.X.); (S.L.); (H.L.); (K.R.); (Y.J.); (Z.W.); (S.L.); (Z.K.); (Z.C.)
- Xi’an Key Laboratory of Special Medicine and Health Engineering, School of Life Sciences, Northwestern Polytechnical University, Xi’an 710072, China
- Research Center for Special Medicine and Health Systems Engineering, School of Life Sciences, Northwestern Polytechnical University, Xi’an 710072, China
- NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi’an 710072, China
- Correspondence: (A.Q.); (L.H.)
| | - Lifang Hu
- Lab for Bone Metabolism, Key Lab for Space Biosciences and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi’an 710072, China; (X.X.); (S.L.); (H.L.); (K.R.); (Y.J.); (Z.W.); (S.L.); (Z.K.); (Z.C.)
- Xi’an Key Laboratory of Special Medicine and Health Engineering, School of Life Sciences, Northwestern Polytechnical University, Xi’an 710072, China
- Research Center for Special Medicine and Health Systems Engineering, School of Life Sciences, Northwestern Polytechnical University, Xi’an 710072, China
- NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi’an 710072, China
- Correspondence: (A.Q.); (L.H.)
| |
Collapse
|
49
|
Chen P, Zhang G, Jiang S, Ning Y, Deng B, Pan X, Liu S, He Y, Zhang L, Wan R, Wu Z, He Q, Yin J, Wang H, Li J. Mechanosensitive Piezo1 in endothelial cells promotes angiogenesis to support bone fracture repair. Cell Calcium 2021; 97:102431. [PMID: 34153657 DOI: 10.1016/j.ceca.2021.102431] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 05/31/2021] [Accepted: 06/01/2021] [Indexed: 12/22/2022]
Abstract
Piezo1, a calcium-permeable non-selective cationic channel that senses mechanical stimulation in multicellular organisms, mediates various biological processes, including angiogenesis. The supply of nutrients and oxygen through newly formed blood vessels at the fractured lesion is critical for bone fracture repair. The elucidation of the underlying mechanisms involved in angiogenesis and bone repair can aid in improving fracture healing. Here, mice with endothelial cell-specific deletion of Piezo1 channels were used to examine the role of Piezo1 in the initiation of fracture healing. The expression and distribution of Piezo1 was explored in the vasculature of the bone. The deletion of endothelial Piezo1 resulted in impaired bone fracture repair, downregulation of calcium-activated proteolytic calpain activity during vascularization, inhibition of osteoblast maturation and ossification, downregulation of phosphorylated PI3K-AKT, and impaired Notch signaling during bone fracture union. These findings indicated that Piezo1 protein is a potential target for enhancing bone regeneration and treating delayed or nonunion bone fractures.
Collapse
Affiliation(s)
- Peng Chen
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Gangyu Zhang
- The First School of Clinical Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Shan Jiang
- The First School of Clinical Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Yile Ning
- Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; The First School of Clinical Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Bo Deng
- Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; The First School of Clinical Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Xianmei Pan
- Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; The First School of Clinical Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Silin Liu
- Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; The First School of Clinical Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Yu He
- Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; The First School of Clinical Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Lei Zhang
- Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; The First School of Clinical Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Rentao Wan
- Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; The First School of Clinical Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Zhiming Wu
- Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; The First School of Clinical Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Qi He
- Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; The First School of Clinical Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Jiang Yin
- The affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou 510095, China
| | - Haibin Wang
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou 510405, China.
| | - Jing Li
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, LS2 9JT, UK.
| |
Collapse
|
50
|
Abstract
Periodontitis is one of the most prevalent epidemics affecting human health and life recently, and exploration of the pathogenesis and treatment of periodontitis has been valued by scholars. In recent years, sclerostin, a new factor on bone resorption and reconstruction caused by inflammation and mechanical stimulation, has been a research hotspot. This article summarizes the researches on sclerostin in periodontitis development in recent years. Among them, sclerostin has been shown to be a critical negative regulator of bone formation, thereby inhibiting bone remodeling in periodontitis development, and is closely associated with tooth movement. Besides, evidence indicates that the removal of sclerostin seems to reasonably protect the alveolar bone from resorption. Regulation of sclerostin expression is a novel, promising treatment for periodontitis and addresses several complications seen with traditional therapies; accordingly, many drugs with similar mechanisms have emerged. Moreover, the application prospect of sclerostin in periodontal therapy combined with orthodontic treatment is another promising approach. There are also a lot of drugs that regulate sclerostin. Anti-sclerostin antibody (Scl-Ab) is the most direct one that inhibits bone resorption caused by sclerostin. At present, drugs that inhibit the expression of sclerostin have been applied to the treatment of diseases such as multiple myeloma and osteoporosis. Therefore, the application of sclerostin in the oral field is just around the corner, which provides a new therapeutic bone regulation strategy in oral and general health.
Collapse
|