1
|
Tandon B, Aguilar Cosme JR, Xue R, Srirussamee K, Aguilar-Tadeo J, Ballestrem C, Blaker JJ, Cartmell SH. Co-stimulation with piezoelectric PVDF films and low intensity pulsed ultrasound enhances osteogenic differentiation. BIOMATERIALS ADVANCES 2025; 173:214283. [PMID: 40086006 DOI: 10.1016/j.bioadv.2025.214283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Revised: 03/06/2025] [Accepted: 03/08/2025] [Indexed: 03/16/2025]
Abstract
Bone tissue engineering has emerged as a promising approach to address the challenges of bone fracture repair and regeneration. The application of external stimuli (mechanical and electrical) can drive specific cellular responses and osteogenic differentiation, leading to the development of more effective treatments. Piezoelectric materials modulate cellular proliferation and osteogenic differentiation under both static (without mechanical stimulation) and dynamic (with mechanical stimulation) conditions, activating distinct gene expression pathways. In this work, we investigate the combinatorial effect of poly (vinylidene fluoride) (PVDF) poled and non-poled films, and low-intensity pulsed ultrasound (LIPUS) on early-stage osteogenic differentiation of mouse pre-osteoblasts. Static culture with PVDF poled films enhanced Runx2 and Col1α1 expression without impacting alkaline phosphatase (ALP) activity. Inhibition of ERK phosphorylation using U0126 in PVDF poled films resulted in a ~ 6-8-fold increase in ALP activity, suggesting the involvement of an alternative pathway in osteogenic differentiation. Dynamic culture with LIPUS generated an electric potential of approximately 500 mV across PVDF films and an electrical field of 0-10 mV mm-1. Co-stimulation led to a ~3-fold increase of ALP activity on stimulated PVDF compared to unstimulated films. This study underscores the potential of piezoelectric materials as non-invasive electrical stimulators to enhance the efficacy of ultrasound-based therapies for bone fracture repair.
Collapse
Affiliation(s)
- Biranche Tandon
- Department of Materials Science, School of Natural Sciences, Faculty of Science and Engineering, The University of Manchester, Manchester, UK
| | - Jose R Aguilar Cosme
- Department of Materials Science, School of Natural Sciences, Faculty of Science and Engineering, The University of Manchester, Manchester, UK; Henry Royce Institute, The University of Manchester, Manchester, UK
| | - Ruikang Xue
- Department of Materials Science, School of Natural Sciences, Faculty of Science and Engineering, The University of Manchester, Manchester, UK; Department of Mechanical Engineering, Faculty of Engineering Science, University College London, London WC1E 7JE, UK
| | - Kasama Srirussamee
- Department of Materials Science, School of Natural Sciences, Faculty of Science and Engineering, The University of Manchester, Manchester, UK; Department of Biomedical Engineering, School of Engineering, KMITL, Bangkok 10520, Thailand
| | - Julio Aguilar-Tadeo
- Department of Materials Science, School of Natural Sciences, Faculty of Science and Engineering, The University of Manchester, Manchester, UK
| | - Christoph Ballestrem
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester M13 9PL, UK
| | - Jonny J Blaker
- Department of Materials Science, School of Natural Sciences, Faculty of Science and Engineering, The University of Manchester, Manchester, UK; Henry Royce Institute, The University of Manchester, Manchester, UK
| | - Sarah H Cartmell
- Department of Materials Science, School of Natural Sciences, Faculty of Science and Engineering, The University of Manchester, Manchester, UK; Henry Royce Institute, The University of Manchester, Manchester, UK.
| |
Collapse
|
2
|
Qin L, Cao H, Liu X, Zhang D, Wu L. Low-Intensity Pulsed Ultrasound Promotes Osteogenesis in Porous Titanium Alloys Through miR-1187/BMP4 Pathway. FASEB J 2025; 39:e70583. [PMID: 40317956 PMCID: PMC12060846 DOI: 10.1096/fj.202403395rr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2024] [Revised: 04/04/2025] [Accepted: 04/18/2025] [Indexed: 05/07/2025]
Abstract
The repair of critical-sized bone defects remains a significant clinical challenge. Low-intensity pulsed ultrasound (LIPUS), in combination with porous titanium alloy (PTi) scaffolds, has emerged as a promising therapeutic strategy. However, its molecular mechanism remains unclear. This study aimed to investigate the role of bone morphogenetic protein 4 (BMP4) and microRNA-1187 (miR-1187) in LIPUS-mediated osteogenesis in PTi scaffolds. In vitro, the expression of BMP4 and miR-1187 in MC3T3-E1 cells following LIPUS stimulation was assessed using quantitative real-time PCR (RT-qPCR), western blotting, ELISA, alkaline phosphatase (ALP) activity assay, and staining techniques. A luciferase reporter assay confirmed BMP4 as a direct target of miR-1187. Functional studies demonstrated that BMP4 overexpression and miR-1187 inhibition promoted osteoblast differentiation, whereas BMP4 knockdown and miR-1187 overexpression suppressed osteogenesis. In vivo, a BMP4 knockdown rat model was established by si-BMP4 injection into mandibular defects and evaluated new bone formation using micro-CT and histological analyses. LIPUS stimulation significantly upregulated BMP4 expression, promoted new bone formation in PTi scaffolds, and partially rescued the inhibitory effects of BMP4 silencing. These findings establish BMP4 as a key regulator in LIPUS-enhanced osteogenesis via miR-1187 suppression. This mechanistic insight supports the combined use of LIPUS and PTi scaffolds for bone defect repair and highlights BMP4 as a potential therapeutic target to further enhance bone regeneration in LIPUS-stimulated scaffold therapies.
Collapse
Affiliation(s)
- Limei Qin
- Department of Prosthodontics, School of StomatologyChina Medical UniversityShenyangChina
| | - Hongjuan Cao
- Department of Prosthodontics, School of StomatologyChina Medical UniversityShenyangChina
| | - Xiaohan Liu
- Department of Prosthodontics, School of StomatologyChina Medical UniversityShenyangChina
| | - Di Zhang
- Department of Prosthodontics, School of StomatologyChina Medical UniversityShenyangChina
| | - Lin Wu
- Department of Prosthodontics, School of StomatologyChina Medical UniversityShenyangChina
| |
Collapse
|
3
|
Bat-Erdene B, He M, Dong J, Li Y, Ta D. Therapeutic Effects of Different Ultrasound Intensity Stimulation on Brown Adipose Tissue for the Treatment of Type 2 Diabetes. ULTRASOUND IN MEDICINE & BIOLOGY 2025; 51:830-840. [PMID: 39924417 DOI: 10.1016/j.ultrasmedbio.2025.01.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 01/10/2025] [Accepted: 01/14/2025] [Indexed: 02/11/2025]
Abstract
Type 2 diabetes (T2D) is a persistent illness that has a high incidence rate. Still, there is no conclusive evidence on effectively improving blood sugar levels in patients through physical therapy. This study examined the regulatory effects of different intensities of low-intensity pulsed ultrasound (LIPUS) on T2D by stimulating brown adipose tissue (BAT). Eight-week-old C57BL/6J mice were divided into six groups (n = 10 per group): Control sham (C-Sham), Control-LIPUS (C-LIPUS), T2D-sham (T2D-Sham), T2D groups treated with LIPUS at spatial average-temporal-average intensity (Isata) of 60mW/cm² (T2D-L-60), 80mW/cm² (T2D-L-80), and 100mW/cm² (T2D-L-100). T2D models were induced by intraperitoneal injection of 40 mg/kg streptozotocin (STZ) three times after 12 wks of high-fat diet (HFD). The T2D-LIPUS group received LIPUS stimulation for 20 minutes per day for 6 weeks. The LIPUS stimulation had a duty cycle of 20%, a frequency of 1 MHz, and Isata of 60mW/cm², 80mW/cm², 100mW/cm². Subsequently, glucose tolerance tests (GTT) and insulin tolerance tests (ITT) were performed, and body fat content in mice was analyzed using nuclear magnetic resonance (NMR). Metabolic changes were monitored using metabolic cages. The results indicated that 80mW/cm² intensity level significantly improved glucose tolerance, insulin sensitivity, and metabolic function after LIPUS exposure. Significant reductions in body fat content and enhanced thermogenesis were observed, highlighting the potential of LIPUS in T2D management. This provides the basis for the dose study of LIPUS in the treatment of T2D.
Collapse
Affiliation(s)
- Badamgarav Bat-Erdene
- Department of Biomedical Engineering, School of Information Science and Technology, Fudan University, Shanghai, China
| | - Min He
- Department of Biomedical Engineering, School of Information Science and Technology, Fudan University, Shanghai, China.
| | - Jingsong Dong
- Department of Biomedical Engineering, School of Information Science and Technology, Fudan University, Shanghai, China
| | - Ying Li
- Department of Biomedical Engineering, School of Information Science and Technology, Fudan University, Shanghai, China
| | - Dean Ta
- Department of Biomedical Engineering, School of Information Science and Technology, Fudan University, Shanghai, China; Academy for Engineering and Technology, Fudan University, Shanghai, China; State Key Laboratory of Integrated Chips and Systems, Fudan University, Shanghai, China; Department of Rehabilitation Medicine, Huashan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
4
|
Li D, Yong Y, Qiao C, Jiang H, Lin J, Wei J, Zhou Y, Li F. Low-Intensity Pulsed Ultrasound Dynamically Modulates the Migration of BV2 Microglia. ULTRASOUND IN MEDICINE & BIOLOGY 2025; 51:494-507. [PMID: 39632209 DOI: 10.1016/j.ultrasmedbio.2024.11.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 10/18/2024] [Accepted: 11/06/2024] [Indexed: 12/07/2024]
Abstract
OBJECTIVE Low-intensity pulsed ultrasound (LIPUS) is a promising modality for neuromodulation. Microglia are the resident immune cells in the brain and their mobility is critical for maintaining brain homeostasis and alleviating neuroimmune pathologies. However, it is unclear whether and how LIPUS modulates microglial migration in physiological conditions. METHODS Here we examined the in vitro effects of LIPUS on the mobility of BV2 microglia by live cell imaging. Single-cell tracing of BV2 microglia migration was analyzed using ImageJ and Chemotaxis and Migration Tool software. Pharmacological manipulation was performed to determine the key molecular players involved in regulating ultrasound-dependent microglia migration. RESULTS We found that the distance of microglial migration was enhanced by LIPUS with increasing acoustic pressure. Removing the extracellular Ca2+ influx or depletion of intracellular Ca2+ stores suppressed ultrasound-enhanced BV2 migration. Furthermore, we found that blocking the reorganization of actin, or suppressing purinergic signaling by application of apyrase or hemi-channel inhibitors, both diminished ultrasound-induced BV2 migration. LIPUS stimulation also enhanced microglial migration in a lipopolysaccharide (LPS)-induced inflammatory environment. CONCLUSION LIPUS promoted microglia migration in both physiological and inflammatory environments. Calcium, cytoskeleton, and purinergic signaling were involved in regulating ultrasound-dependent microglial mobility. Our study reveals the biomechanical impact of ultrasound on microglial migration and highlights the potential of using ultrasound-based tools for modulation of microglial function.
Collapse
Affiliation(s)
- Dandan Li
- Institute of Biomedical Engineering, Shenzhen Bay Laboratory, Shenzhen, China
| | - Yu Yong
- Institute of Biomedical Engineering, Shenzhen Bay Laboratory, Shenzhen, China
| | - Chaofeng Qiao
- Institute of Biomedical Engineering, Shenzhen Bay Laboratory, Shenzhen, China; School of Basic Medical Sciences, Beihua University, Jilin City, China
| | - Hao Jiang
- Institute of Biomedical Engineering, Shenzhen Bay Laboratory, Shenzhen, China
| | - Jiawei Lin
- Institute of Biomedical Engineering, Shenzhen Bay Laboratory, Shenzhen, China
| | - Jianpeng Wei
- Institute of Biomedical Engineering, Shenzhen Bay Laboratory, Shenzhen, China
| | - Yufeng Zhou
- Chongqing Medical University, State Key Laboratory of Ultrasound in Medicine and Engineering, Chongqing City, China
| | - Fenfang Li
- Institute of Biomedical Engineering, Shenzhen Bay Laboratory, Shenzhen, China.
| |
Collapse
|
5
|
Park J, Na YC, Lee J, Seo Y, Kim H, Han S, Song BW, Chang WS. Role of P 2 × 7 receptor during focused ultrasound induced blood brain barrier modulation. Sci Rep 2025; 15:965. [PMID: 39762398 PMCID: PMC11704064 DOI: 10.1038/s41598-024-83913-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Accepted: 12/18/2024] [Indexed: 01/11/2025] Open
Abstract
Although low-intensity focused ultrasound (LiFUS) with microbubbles is used to temporally open the blood-brain barrier (BBB), the underlying mechanism is not fully understood. This study aimed to analyze BBB-related alterations in the brain microenvironment after LiFUS, with a focus on the involvement of the purinergic P2 × 7 receptor. Sprague-Dawley rats were sonicated with LiFUS at 0.3 MPa energy. The impact of LiFUS on the P2 × 7 receptor and inflammatory-related proteins, including NLRP3 and interleukin-1β, was analyzed through western blotting. The BBB-associated tight junction proteins, zonula occludens-1 (ZO-1) and occludin, were also analyzed. BBB permeability was assessed by quantifying the amount of Evans blue dye penetration using spectrophotometry. Furthermore, the safety of the sonication procedure was verified via terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) assay and hematoxylin and eosin staining. Substantial increases in the P2 × 7 receptor and its downstream signaling were confirmed after sonicating the BBB with LiFUS for 1 h (p < 0.05). Conversely, for tight junction proteins, the lowest expression was observed at 1 h (p < 0.001). Both responses were normalized back to the original state over time. No evidence of brain damage was observed during the procedure. Furthermore, the P2 × 7 receptor antagonist-injected group showed reduced Evans blue dye penetration compared to that 1 h after FUS, indicating a mitigated impact of LiFUS on the BBB. Herein, we elucidate the underlying mechanism by which LiFUS affects the BBB, with a focus on the involvement of the P2 × 7 receptor. Our findings demonstrate that the extent of BBB opening varies upon the regulation of the P2 × 7 receptor. This study provides valuable insights into the mechanisms underlying BBB modulation through LiFUS, thereby laying the foundation for expanding its applications.
Collapse
Affiliation(s)
- Junwon Park
- Department of Neurosurgery and Brain Research Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Young Cheol Na
- Department of Neurosurgery and Brain Research Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
- Department of Neurosurgery, Catholic Kwandong University College of Medicine, International St Mary's Hospital, Incheon Metropolitan City, Republic of Korea
| | | | - Younghee Seo
- Department of Neurosurgery and Brain Research Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Hojin Kim
- Department of Medical Science, College of Medicine, Catholic Kwandong University, Gangneung-si, Gangwon-do, Republic of Korea
| | - Sangheon Han
- Department of Neurosurgery and Brain Research Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Byeong-Wook Song
- Department of Medical Science, College of Medicine, Catholic Kwandong University, Gangneung-si, Gangwon-do, Republic of Korea.
| | - Won Seok Chang
- Department of Neurosurgery and Brain Research Institute, Yonsei University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
6
|
Marcotulli M, Barbetta A, Scarpa E, Bini F, Marinozzi F, Ruocco G, Casciola CM, Scognamiglio C, Carugo D, Cidonio G. Jingle Cell Rock: Steering Cellular Activity With Low-Intensity Pulsed Ultrasound (LIPUS) to Engineer Functional Tissues in Regenerative Medicine. ULTRASOUND IN MEDICINE & BIOLOGY 2024; 50:1973-1986. [PMID: 39289118 DOI: 10.1016/j.ultrasmedbio.2024.08.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 08/15/2024] [Accepted: 08/23/2024] [Indexed: 09/19/2024]
Abstract
Acoustic manipulation or perturbation of biological soft matter has emerged as a promising clinical treatment for a number of applications within regenerative medicine, ranging from bone fracture repair to neuromodulation. The potential of ultrasound (US) endures in imparting mechanical stimuli that are able to trigger a cascade of molecular signals within unscathed cells. Particularly, low-intensity pulsed ultrasound (LIPUS) has been associated with bio-effects such as activation of specific cellular pathways and alteration of cell morphology and gene expression, the extent of which can be modulated by fine tuning of LIPUS parameters including intensity, frequency and exposure time. Although the molecular mechanisms underlying LIPUS are not yet fully elucidated, a number of studies clearly define the modulation of specific ultrasonic parameters as a means to guide the differentiation of a specific set of stem cells towards adult and fully differentiated cell types. Herein, we outline the applications of LIPUS in regenerative medicine and the in vivo and in vitro studies that have confirmed the unbounded clinical potential of this platform. We highlight the latest developments aimed at investigating the physical and biological mechanisms of action of LIPUS, outlining the most recent efforts in using this technology to aid tissue engineering strategies for repairing tissue or modelling specific diseases. Ultimately, we detail tissue-specific applications harnessing LIPUS stimuli, offering insights over the engineering of new constructs and therapeutic modalities. Overall, we aim to lay the foundation for a deeper understanding of the mechanisms governing LIPUS-based therapy, to inform the development of safer and more effective tissue regeneration strategies in the field of regenerative medicine.
Collapse
Affiliation(s)
- Martina Marcotulli
- 3D Microfluidic Bioprinting Lab, Center for Life Nano- & Neuro-Science (CLN2S), Italian Institute of Technology (IIT), Rome, Italy; Department of Biochemical Sciences, Sapienza University of Rome, Rome, Italy; Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences (NDORMS), University of Oxford, Oxford, UK
| | - Andrea Barbetta
- Department of Chemistry, Sapienza University of Rome, Rome, Italy
| | - Edoardo Scarpa
- Infection Dynamics Laboratory, Department of Pharmaceutical Sciences, University of Milan, Milan, Italy; National Institute of Molecular Gentics (INGM), Milan, Italy; Bone and Joint Research Group, Centre for Human Development, Stem Cells and Regeneration, Institute of Developmental Sciences, University of Southampton, Southampton, UK
| | - Fabiano Bini
- Department of Mechanical and Aerospace Engineering (DIMA), Sapienza University of Rome, Rome, Italy
| | - Franco Marinozzi
- Department of Mechanical and Aerospace Engineering (DIMA), Sapienza University of Rome, Rome, Italy
| | - Giancarlo Ruocco
- 3D Microfluidic Bioprinting Lab, Center for Life Nano- & Neuro-Science (CLN2S), Italian Institute of Technology (IIT), Rome, Italy
| | - Carlo Massimo Casciola
- Department of Mechanical and Aerospace Engineering (DIMA), Sapienza University of Rome, Rome, Italy
| | - Chiara Scognamiglio
- 3D Microfluidic Bioprinting Lab, Center for Life Nano- & Neuro-Science (CLN2S), Italian Institute of Technology (IIT), Rome, Italy
| | - Dario Carugo
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences (NDORMS), University of Oxford, Oxford, UK
| | - Gianluca Cidonio
- 3D Microfluidic Bioprinting Lab, Center for Life Nano- & Neuro-Science (CLN2S), Italian Institute of Technology (IIT), Rome, Italy; Bone and Joint Research Group, Centre for Human Development, Stem Cells and Regeneration, Institute of Developmental Sciences, University of Southampton, Southampton, UK; Department of Mechanical and Aerospace Engineering (DIMA), Sapienza University of Rome, Rome, Italy.
| |
Collapse
|
7
|
Müller WEG, Neufurth M, Wang S, Schröder HC, Wang X. Polyphosphate Nanoparticles: Balancing Energy Requirements in Tissue Regeneration Processes. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2309528. [PMID: 38470207 DOI: 10.1002/smll.202309528] [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: 10/20/2023] [Revised: 01/29/2024] [Indexed: 03/13/2024]
Abstract
Nanoparticles of a particular, evolutionarily old inorganic polymer found across the biological kingdoms have attracted increasing interest in recent years not only because of their crucial role in metabolism but also their potential medical applicability: it is inorganic polyphosphate (polyP). This ubiquitous linear polymer is composed of 10-1000 phosphate residues linked by high-energy anhydride bonds. PolyP causes induction of gene activity, provides phosphate for bone mineralization, and serves as an energy supplier through enzymatic cleavage of its acid anhydride bonds and subsequent ATP formation. The biomedical breakthrough of polyP came with the development of a successful fabrication process, in depot form, as Ca- or Mg-polyP nanoparticles, or as the directly effective polymer, as soluble Na-polyP, for regenerative repair and healing processes, especially in tissue areas with insufficient blood supply. Physiologically, the platelets are the main vehicles for polyP nanoparticles in the circulating blood. To be biomedically active, these particles undergo coacervation. This review provides an overview of the properties of polyP and polyP nanoparticles for applications in the regeneration and repair of bone, cartilage, and skin. In addition to studies on animal models, the first successful proof-of-concept studies on humans for the healing of chronic wounds are outlined.
Collapse
Affiliation(s)
- Werner E G Müller
- ERC Advanced Investigator Grant Research Group at the Institute for Physiological Chemistry, University Medical Center of the Johannes Gutenberg University, Duesbergweg 6, D-55128, Mainz, Germany
| | - Meik Neufurth
- ERC Advanced Investigator Grant Research Group at the Institute for Physiological Chemistry, University Medical Center of the Johannes Gutenberg University, Duesbergweg 6, D-55128, Mainz, Germany
| | - Shunfeng Wang
- ERC Advanced Investigator Grant Research Group at the Institute for Physiological Chemistry, University Medical Center of the Johannes Gutenberg University, Duesbergweg 6, D-55128, Mainz, Germany
| | - Heinz C Schröder
- ERC Advanced Investigator Grant Research Group at the Institute for Physiological Chemistry, University Medical Center of the Johannes Gutenberg University, Duesbergweg 6, D-55128, Mainz, Germany
| | - Xiaohong Wang
- ERC Advanced Investigator Grant Research Group at the Institute for Physiological Chemistry, University Medical Center of the Johannes Gutenberg University, Duesbergweg 6, D-55128, Mainz, Germany
| |
Collapse
|
8
|
Tang L, Wu T, Li J, Yu Y, Ma Z, Sun L, Ta D, Fan X. Study on Synergistic Effects of Nanohydroxyapatite/High-Viscosity Carboxymethyl Cellulose Scaffolds Stimulated by LIPUS for Bone Defect Repair of Rats. ACS Biomater Sci Eng 2024; 10:1018-1030. [PMID: 38289029 DOI: 10.1021/acsbiomaterials.3c01381] [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] [Indexed: 02/13/2024]
Abstract
Despite the self-healing capacity of bone, the regeneration of critical-size bone defects remains a major clinical challenge. In this study, nanohydroxyapatite (nHAP)/high-viscosity carboxymethyl cellulose (hvCMC, 6500 mPa·s) scaffolds and low-intensity pulsed ultrasound (HA-LIPUS) were employed to repair bone defects. First, hvCMC was prepared from ramie fiber, and the degree of substitution (DS), purity, and content of NaCl of hvCMC samples were 0.91, 99.93, and 0.017%, respectively. Besides, toxic metal contents were below the permissible limits for pharmaceutically used materials. Our results demonstrated that the hvCMC is suitable for pharmaceutical use. Second, nHAP and hvCMC were employed to prepare scaffolds by freeze-drying. The results indicated that the scaffolds were porous, and the porosity was 35.63 ± 3.52%. Subsequently, the rats were divided into four groups (n = 8) randomly: normal control (NC), bone defect (BD), bone defect treated with nHAP/hvCMC scaffolds (HA), and bone defect treated with nHAP/hvCMC scaffolds and stimulated by LIPUS (HA-LIPUS). After drilling surgery, nHAP/hvCMC scaffolds were implanted in the defect region of HA and HA-LIPUS rats. Meanwhile, HA-LIPUS rats were treated by LIPUS (1.5 MHz, 80 mW cm-2) irradiation for 2 weeks. Compared with BD rats, the maximum load and bone mineral density of HA-LIPUS rats were increased by 20.85 and 51.97%, respectively. The gene and protein results indicated that nHAP/hvCMC scaffolds and LIPUS promoted the bone defect repair and regeneration of rats significantly by activating Wnt/β-catenin and inhibiting OPG/RANKL signaling pathways. Overall, compared with BD rats, nHAP/hvCMC scaffolds and LIPUS promoted bone defect repair significantly. Furthermore, the research results also indicated that there are synergistic effects for bone defect repair between the nHAP/hvCMC scaffolds and LIPUS.
Collapse
Affiliation(s)
- Liang Tang
- Institute of Sports Biology, Shaanxi Normal University, Xi'an 710119, China
| | - Tianpei Wu
- Institute of Sports Biology, Shaanxi Normal University, Xi'an 710119, China
| | - Jiaxiang Li
- Institute of Sports Biology, Shaanxi Normal University, Xi'an 710119, China
| | - Yanan Yu
- Institute of Sports Biology, Shaanxi Normal University, Xi'an 710119, China
| | - Zhanke Ma
- Institute of Sports Biology, Shaanxi Normal University, Xi'an 710119, China
| | - Lijun Sun
- Institute of Sports Biology, Shaanxi Normal University, Xi'an 710119, China
| | - Dean Ta
- Center for Biomedical Engineering, School of Information Science and Technology, Fudan University, Shanghai 200433, China
- Department of Rehabilitation Medicine, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Xiushan Fan
- Institute of Sports Biology, Shaanxi Normal University, Xi'an 710119, China
| |
Collapse
|
9
|
Vaddamanu SK, Alhamoudi FH, Vyas R, Gurumurthy V, Siurkel Y, Cicciù M, Minervini G. Attenuation of orthodontically induced inflammatory root resorption by using low-intensity pulsed ultrasound as a therapeutic modality- a systematic review. BMC Oral Health 2024; 24:67. [PMID: 38200481 PMCID: PMC10782536 DOI: 10.1186/s12903-023-03741-1] [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: 10/30/2023] [Accepted: 12/06/2023] [Indexed: 01/12/2024] Open
Abstract
Ultrasound is an effective tool for both diagnostic and therapeutic applications. As an imaging tool, ultrasound has mostly been used for real-time noninvasive diagnostic imaging. As ultrasound propagates through a material, a reflected radio-frequency (RF) signal is generated when encountering a mismatch in acoustic impedance. While traditionally recognized for its diagnostic imaging capabilities, the application of ultrasound has broadened to encompass therapeutic interventions, most notably in the form of Low-Intensity Pulsed Ultrasound (LIPUS). Low-Intensity Pulsed Ultrasound (LIPUS) is a form of mechanical energy transmitted transcutaneously by high-frequency acoustic pressure waves. The intensity of LIPUS (30 mW/cm2) is within the range of ultrasound intensities used for diagnostic purposes (1-50 mW/cm2) and is regarded as non-thermal, non-destructive, permeating living tissues and triggering a cascade of biochemical responses at the cellular level. The LIPUS device produces a 200 µs burst of 1.5 MHz acoustic sine waves, that repeats at a modulation frequency of 1 kHz and provides a peak pressure of 30 mW/cm2. Low-intensity pulsed ultrasound (LIPUS) forms one of the currently available non-invasive healing-enhancing devices besides electro-stimulation (pulsed electro-magnetic field, PEMF). This modality has been leveraged to enhance drug delivery, expedite injury recovery, improve muscle mobility, alleviate joint stiffness and muscle pain, and enhance bone fracture healing. Although LIPUS has been embraced within various medical disciplines, its integration into standard dental practices is still in its nascent stages, signifying an unexplored frontier with potentially transformative implications. Low-intensity pulsed ultrasound (LIPUS) has emerged as an attractive adjuvant therapy in various dental procedures, such as orthodontic treatment and maxillary sinus augmentation. Its appeal lies in its simplicity and non-invasive nature, positioning LIPUS as a promising avenue for clinical innovation. One particular area of interest is orthodontically induced inflammatory root resorption (OIIRR), an oftenunavoidable outcome of the orthodontic intervention, resulting in the permanent loss of root structure. Notably, OIIRR is the second most common form of root resorption (RR), surpassed only by root resorption related to pulpal infection. Given the high prevalence and potential long-term consequences of OIIRR, this literature review seeks to evaluate the efficacy of LIPUS as a therapeutic approach, with an emphasis on assessing its capacity to reduce the severity of OIIRR to a level of clinical significance. To conduct this systematic review, a comprehensive automated literature search was executed across multiple databases, including MEDLINE, Embase, PsycINFO, Web of Knowledge, Scopus, CINAHL, LILACS, SciELO, Cochrane, PubMed, trials registries, 3ie, and Google Scholar. Both forward and backward citation tracking was employed, encompassing studies published from database inception through January 2009 to April 2023. The review focused on randomized controlled trials (RCTs) that specifically evaluated the effects of low-intensity pulsed ultrasound therapy on orthodontically induced inflammatory root resorption (OIIRR), without restrictions of publication date. A stringent selection criterion was applied, and only studies demonstrating high levels of statistical significance were included. Ultimately, fourteen studies met the inclusion criteria and were subjected to further analysis. The overall quality of the included randomized controlled trials (RCTs) was rigorously assessed utilizing the Grading of Recommendations Assessment, Development, and Evaluation (GRADE) approach. This analysis revealed certain methodological limitations that posed challenges in drawing definitive conclusions from the available evidence. Despite these constraints, the review offers invaluable insights that can inform and guide future research. Specifically, it delineates recommendations for targeted populations, necessary interventions, appropriate outcome measures, suitable study designs, and essential infrastructure to facilitate further investigations. The synthesis of these insights aims to enhance the development and application of low-intensity pulsed ultrasound therapy within the field of dentistry, thereby contributing to improved patient outcomes.
Collapse
Affiliation(s)
- Sunil Kumar Vaddamanu
- Department of Dental Technology, College of Applied Medical Sciences, King Khalid University, Abha, 62529, Saudi Arabia.
| | - Fahad Hussain Alhamoudi
- Department of Dental Technology, College of Applied Medical Sciences, King Khalid University, Abha, 62529, Saudi Arabia
| | - Rajesh Vyas
- Department of Dental Technology, College of Applied Medical Sciences, King Khalid University, Abha, 62529, Saudi Arabia
| | - Vishwanath Gurumurthy
- Department of Dental Technology, College of Applied Medical Sciences, King Khalid University, Abha, 62529, Saudi Arabia
| | - Yulia Siurkel
- International European University School of Medicine, Akademika Hlushkova Ave, 42В, Kyiv, 03187, Ukraine.
| | - Marco Cicciù
- Department of Biomedical and Surgical and Biomedical Sciences, Catania University, Catania, 95123, Italy
| | - Giuseppe Minervini
- Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences (SIMATS), Saveetha University, Chennai, Tamil Nadu, India.
- Multidisciplinary Department of Medical-Surgical and Dental Specialties, University of Campania Luigi Vanvitelli, Caserta, 81100, Italy.
| |
Collapse
|
10
|
Titanium dioxide nanotubes increase purinergic receptor P2Y6 expression and activate its downstream PKCα-ERK1/2 pathway in bone marrow mesenchymal stem cells under osteogenic induction. Acta Biomater 2023; 157:670-682. [PMID: 36442823 DOI: 10.1016/j.actbio.2022.11.045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 10/25/2022] [Accepted: 11/21/2022] [Indexed: 11/27/2022]
Abstract
Titanium dioxide (TiO2) nanotubes can improve the osseointegration of pure titanium implants, but this exact mechanism has not been fully elucidated. The purinergic receptor P2Y6 is expressed in bone marrow mesenchymal stem cells (BMSCs) and participates in the regulation of bone metabolism. However, it is unclear as to whether P2Y6 is involved in the osteogenic differentiation of BMSCs induced by TiO2 nanotubes. TiO2 nanotubes were prepared on the surface of titanium specimens using the anodizing method and characterized their features. Quantitative reverse transcriptase polymerase chain reaction and western blotting were used to detect the expression of P2Y6, markers of osteogenic differentiation, and PKCα-ERK1/2. A rat femoral defect model was established to evaluate the osseointegration effect of TiO2 nanotubes combined with P2Y6 agonists. The results showed that the average inner diameter of the TiO2 nanotubes increased with an increase in voltage (voltage range of 30-90V), and the expression of P2Y6 in BMSCs could be upregulated by TiO2 nanotubes in osteogenic culture. Inhibition of P2Y6 expression partially inhibited the osteogenic effect of TiO2 nanotubes and downregulated the activity of the PKCα-ERK1/2 pathway. When using in vitro and in vivo experiments, the osteogenic effect of TiO2 nanotubes when combined with P2Y6 agonists was more pronounced. TiO2 nanotubes promoted the P2Y6 expression of BMSCs during osteogenic differentiation and promoted osteogenesis by activating the PKCα-ERK1/2 pathway. The combined application of TiO2 nanotubes and P2Y6 agonists may be an effective new strategy to improve the osseointegration of titanium implants. STATEMENT OF SIGNIFICANCE: Titanium dioxide (TiO2) nanotubes can improve the osseointegration of pure titanium implants, but this exact mechanism has not been fully elucidated. The purinergic receptor P2Y6 is expressed in bone marrow mesenchymal stem cells (BMSCs) and participates in the regulation of bone metabolism. However, it is unclear as to whether P2Y6 is involved in the osteogenic differentiation of BMSCs induced by TiO2 nanotubes. For the first time, this study revealed the relationship between TiO2 nanotubes and purine receptor P2Y6, and further explored its mode of action, which may provide clues as to the regulatory role of TiO2 nanotubes on osteogenic differentiation of BMSCs. These findings will help to develop novel methods for guiding material design and biosafety evaluation of nano implants.
Collapse
|
11
|
Chang X, Xu S, Zhang H. Regulation of bone health through physical exercise: Mechanisms and types. Front Endocrinol (Lausanne) 2022; 13:1029475. [PMID: 36568096 PMCID: PMC9768366 DOI: 10.3389/fendo.2022.1029475] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Accepted: 11/15/2022] [Indexed: 12/12/2022] Open
Abstract
Osteoporosis, characterized by bone mineral density reduction, bone mass loss, increased bone fragility, and propensity to fractures, is a common disease in older individuals and one of the most serious health problems worldwide. The imbalance between osteoblasts and osteoclasts results in the predominance of bone resorption and decreased bone formation. In recent years, it has been found that regular and proper exercise not only helps prevent the occurrence of osteoporosis but also adds benefits to osteoporosis therapy; accordingly, bone homeostasis is closely associated with mechanical stress and the intricate crosstalk between osteoblasts and osteoclasts. In this review, we summarize the mechanisms of exercise on osteoporosis and provide new proposals for the prevention and treatment of osteoporosis.
Collapse
Affiliation(s)
- Xinyu Chang
- Department of Traumatic Orthopedics, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Sheng Xu
- National Key Laboratory of Medical Immunology and Institute of Immunology, Institute of Immunology, Naval Medical University, Shanghai, China
| | - Hao Zhang
- Department of Traumatic Orthopedics, the First Affiliated Hospital of Naval Medical University, Naval Medical University, Shanghai, China
| |
Collapse
|
12
|
da Silva ANG, de Oliveira JRS, Madureira ÁNDM, Lima WA, Lima VLDM. Biochemical and Physiological Events Involved in Responses to the Ultrasound Used in Physiotherapy: A Review. ULTRASOUND IN MEDICINE & BIOLOGY 2022; 48:2417-2429. [PMID: 36115728 DOI: 10.1016/j.ultrasmedbio.2022.07.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 07/18/2022] [Accepted: 07/19/2022] [Indexed: 06/15/2023]
Abstract
Therapeutic ultrasound (TUS) is the ultrasound modality widely used in physical therapy for the treatment of acute and chronic injuries of various biological tissues. Its thermal and mechanical effects modify the permeability of the plasma membrane, the flow of ions and molecules and cell signaling and, in this way, promote the cascade of physiological events that culminate in the repair of injuries. This article is a review of the biochemical and physiological effects of TUS with parameters commonly used by physical therapists. Integrins can translate the mechanical signal of the TUS into a cellular biochemical signal for protein synthesis and modification of the active site of enzymes, so cell function and metabolism are modified. TUS also alters the permeability of the plasma membrane, allowing the influx of ions and molecules that modulate the cellular electrochemical signaling pathways. With biochemical and electrochemical signals tampered with, the cellular response to damage is then modified or enhanced. Greater release of pro-inflammatory factors, cytokines and growth factors, increased blood flow and activation of protein kinases also seem to be involved in the therapeutic response of TUS. Although a vast number of publications describe the mechanisms by which TUS can interact with the biological system, little is known about the metabolic possibilities of TUS because of the lack of standardization in its application.
Collapse
Affiliation(s)
- Ayala Nathaly Gomes da Silva
- Laboratório de Lipídios e Aplicaçães de Biomoléculas em Doenças Prevalentes e Negligenciadas, Universidade Federal de Pernambuco, Recife, Brazil
| | - João Ricardhis Saturnino de Oliveira
- Laboratório de Lipídios e Aplicaçães de Biomoléculas em Doenças Prevalentes e Negligenciadas, Universidade Federal de Pernambuco, Recife, Brazil
| | - Álvaro Nóbrega de Melo Madureira
- Laboratório de Lipídios e Aplicaçães de Biomoléculas em Doenças Prevalentes e Negligenciadas, Universidade Federal de Pernambuco, Recife, Brazil
| | - Wildberg Alencar Lima
- Laboratório de Lipídios e Aplicaçães de Biomoléculas em Doenças Prevalentes e Negligenciadas, Universidade Federal de Pernambuco, Recife, Brazil
| | - Vera Lúcia de Menezes Lima
- Laboratório de Lipídios e Aplicaçães de Biomoléculas em Doenças Prevalentes e Negligenciadas, Universidade Federal de Pernambuco, Recife, Brazil.
| |
Collapse
|
13
|
Dsouza C, Moussa MS, Mikolajewicz N, Komarova SV. Extracellular ATP and its derivatives provide spatiotemporal guidance for bone adaptation to wide spectrum of physical forces. Bone Rep 2022; 17:101608. [PMID: 35992507 PMCID: PMC9385560 DOI: 10.1016/j.bonr.2022.101608] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 07/27/2022] [Accepted: 07/29/2022] [Indexed: 11/17/2022] Open
Abstract
ATP is a ubiquitous intracellular molecule critical for cellular bioenergetics. ATP is released in response to mechanical stimulation through vesicular release, small tears in cellular plasma membranes, or when cells are destroyed by traumatic forces. Extracellular ATP is degraded by ecto-ATPases to form ADP and eventually adenosine. ATP, ADP, and adenosine signal through purinergic receptors, including seven P2X ATP-gated cation channels, seven G-protein coupled P2Y receptors responsive to ATP and ADP, and four P1 receptors stimulated by adenosine. The goal of this review is to build a conceptual model of the role of different components of this complex system in coordinating cellular responses that are appropriate to the degree of mechanical stimulation, cell proximity to the location of mechanical injury, and time from the event. We propose that route and amount of ATP release depend on the scale of mechanical forces, ranging from vesicular release of small ATP boluses upon membrane deformation, to leakage of ATP through resealable plasma membrane tears, to spillage of cellular content due to destructive forces. Correspondingly, different P2 receptors responsive to ATP will be activated according to their affinity at the site of mechanical stimulation. ATP is a small molecule that readily diffuses through the environment, bringing the signal to the surrounding cells. ATP is also degraded to ADP which can stimulate a distinct set of P2 receptors. We propose that depending on the magnitude of mechanical forces and distance from the site of their application, ATP/ADP profiles will be different, allowing the relay of information about tissue level injury and proximity. Lastly, ADP is degraded to adenosine acting via its P1 receptors. The presence of large amounts of adenosine without ATP, indicates that an active source of ATP release is no longer present, initiating the transition to the recovery phase. This model consolidates the knowledge regarding the individual components of the purinergic system into a conceptual framework of choreographed responses to physical forces. Cellular bioenergetic molecule ATP is released when cell is mechanically stimulated. ATP release is proportional to the amount of cellular damage. ATP diffusion and transformation to ADP indicates the proximity to the damage. Purinergic receptors form a network choreographing cell response to physical forces. Complete transformation of ATP to adenosine initiates the recovery phase.
Collapse
Affiliation(s)
- Chrisanne Dsouza
- Department of Experimental Surgery, McGill University, Montreal, QC H3G 1A4, Canada
- Shriners Hospitals for Children- Canada, Montreal, QC H4A 0A9, Canada
| | - Mahmoud S. Moussa
- Shriners Hospitals for Children- Canada, Montreal, QC H4A 0A9, Canada
- Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montreal, QC H3A 1G1, Canada
| | - Nicholas Mikolajewicz
- Shriners Hospitals for Children- Canada, Montreal, QC H4A 0A9, Canada
- Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montreal, QC H3A 1G1, Canada
| | - Svetlana V. Komarova
- Department of Experimental Surgery, McGill University, Montreal, QC H3G 1A4, Canada
- Shriners Hospitals for Children- Canada, Montreal, QC H4A 0A9, Canada
- Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montreal, QC H3A 1G1, Canada
- Corresponding author.
| |
Collapse
|
14
|
Tang L, Wu T, Zhou Y, Zhong Y, Sun L, Guo J, Fan X, Ta D. Study on synergistic effects of carboxymethyl cellulose and LIPUS for bone tissue engineering. Carbohydr Polym 2022; 286:119278. [DOI: 10.1016/j.carbpol.2022.119278] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 01/28/2022] [Accepted: 02/18/2022] [Indexed: 02/07/2023]
|
15
|
Mg.ATP-decorated ultrafine magnetic nanofibers: A bone scaffold with high osteogenic and antibacterial properties in the presence of an electromagnetic field. Colloids Surf B Biointerfaces 2021; 210:112256. [PMID: 34875469 DOI: 10.1016/j.colsurfb.2021.112256] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2021] [Revised: 11/27/2021] [Accepted: 11/29/2021] [Indexed: 12/27/2022]
Abstract
In this study, ultrafine magnetic nanofibers were developed for bone regeneration purposes. Nanofibers were acquired by electrospinning using a two-component nanofiber matrix (CP: chitosan (Cs) and polyvinyl alcohol (PVA)) containing different concentrations of succinate conjugated-magnetic hydroxyapatite nanocomposites (SMHA). Hybrid nanofibers (CP&SMHA) containing 5 mg ml-1 of SMHA nanocomposite showed well-defined properties in terms of physicochemical properties and cell behavior. Then, they were modified with adenosine 5'-triphosphate (ATP) and Mg2+ ions. The initial adhesion of mesenchymal stem cells and their proliferation rate on the surface of modified nanofibers (Mg.ATP.CP&SMHA) were significantly increased as compared to those of bare nanofibers. Analysis of common osteogenic markers such as alkaline phosphatase activity and the expression of Runt-related transcription factor 2 and osteocalcin confirmed the osteogenic efficacy enhancement of CP&SMHA nanofibers when they were functionalized with ATP and Mg2+. The utilization of the antagonist of purine receptor, P2X7, revealed that this receptor has a major role in the osteogenesis process induced by Mg.ATP.CP&SMHA. Moreover, the results showed that cell adhesion, proliferation, and differentiation improved as nanofibers were under the influence of the electromagnetic field (EMF), displaying synergistic effects in the process of bone formation. Mg.ATP.CP&SMHA also showed an antibacterial effect against gram-negative and gram-positive bacteria, Escherichia coli and Staphylococcus aureus, respectively. Considering the high osteogenic potential and antibacterial activity of Mg.ATP.CP&SMHA nanofibers particularly in combination with EMF, it can serve as a great candidate for use in bone tissue engineering applications.
Collapse
|
16
|
Chen J, Li S, Jiao Y, Li J, Li Y, Hao YL, Zuo Y. In Vitro Study on the Piezodynamic Therapy with a BaTiO 3-Coating Titanium Scaffold under Low-Intensity Pulsed Ultrasound Stimulation. ACS APPLIED MATERIALS & INTERFACES 2021; 13:49542-49555. [PMID: 34610736 DOI: 10.1021/acsami.1c15611] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
To solve the poor sustainability of electroactive stimulation in clinical therapy, a strategy of combining a piezoelectric BaTiO3-coated Ti6Al4V scaffold and low-intensity pulsed ultrasound (LIPUS) was unveiled and named here as piezodynamic therapy. Thus, cell behavior could be regulated phenomenally by force and electricity simultaneously. First, BaTiO3 was deposited uniformly on the surface of the three-dimensional (3D) printed porous Ti6Al4V scaffold, which endowed the scaffold with excellent force-electricity responsiveness under pulsed ultrasound exposure. The results of live/dead staining, cell scanning electron microscopy, and F-actin staining showed that cells had better viability, better pseudo-foot adhesion, and more muscular actin bundles when they underwent the piezodynamic effect of ultrasound and piezoelectric coating. This piezodynamic therapy activated more mitochondria at the initial stage that intervened in the cell cycle by promoting cells' proliferation and weakened the apoptotic damage. The quantitative real-time polymerase chain reaction data further confirmed that the costimulation of the ultrasound and the piezoelectric scaffolds could trigger adequate current to upregulated the expression of osteogenic-related genes. The continuous electric cues could be generated by the BaTiO3-coated scaffold and intermittent LIPUS stimulation; thereon, more efficient bone healing would be promoted by piezodynamic therapy in future treatment.
Collapse
Affiliation(s)
- Jie Chen
- Research Center for Nano Biomaterials, and Analytical & Testing Center, Sichuan University, Chengdu 610064, People's Republic of China
| | - Shujun Li
- Shenyang National Laboratory for Materials Science, Institute of Metal Research, Chinese Academy of Sciences, 72 Wenhua Road, Shenyang 110016, People's Republic of China
| | - Yilai Jiao
- Shenyang National Laboratory for Materials Science, Institute of Metal Research, Chinese Academy of Sciences, 72 Wenhua Road, Shenyang 110016, People's Republic of China
| | - Jidong Li
- Research Center for Nano Biomaterials, and Analytical & Testing Center, Sichuan University, Chengdu 610064, People's Republic of China
| | - Yubao Li
- Research Center for Nano Biomaterials, and Analytical & Testing Center, Sichuan University, Chengdu 610064, People's Republic of China
| | - Yu-Lin Hao
- Shenyang National Laboratory for Materials Science, Institute of Metal Research, Chinese Academy of Sciences, 72 Wenhua Road, Shenyang 110016, People's Republic of China
| | - Yi Zuo
- Research Center for Nano Biomaterials, and Analytical & Testing Center, Sichuan University, Chengdu 610064, People's Republic of China
| |
Collapse
|
17
|
Osteocytes as main responders to low-intensity pulsed ultrasound treatment during fracture healing. Sci Rep 2021; 11:10298. [PMID: 33986415 PMCID: PMC8119462 DOI: 10.1038/s41598-021-89672-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Accepted: 04/29/2021] [Indexed: 12/11/2022] Open
Abstract
Ultrasound stimulation is a type of mechanical stress, and low-intensity pulsed ultrasound (LIPUS) devices have been used clinically to promote fracture healing. However, it remains unclear which skeletal cells, in particular osteocytes or osteoblasts, primarily respond to LIPUS stimulation and how they contribute to fracture healing. To examine this, we utilized medaka, whose bone lacks osteocytes, and zebrafish, whose bone has osteocytes, as in vivo models. Fracture healing was accelerated by ultrasound stimulation in zebrafish, but not in medaka. To examine the molecular events induced by LIPUS stimulation in osteocytes, we performed RNA sequencing of a murine osteocytic cell line exposed to LIPUS. 179 genes reacted to LIPUS stimulation, and functional cluster analysis identified among them several molecular signatures related to immunity, secretion, and transcription. Notably, most of the isolated transcription-related genes were also modulated by LIPUS in vivo in zebrafish. However, expression levels of early growth response protein 1 and 2 (Egr1, 2), JunB, forkhead box Q1 (FoxQ1), and nuclear factor of activated T cells c1 (NFATc1) were not altered by LIPUS in medaka, suggesting that these genes are key transcriptional regulators of LIPUS-dependent fracture healing via osteocytes. We therefore show that bone-embedded osteocytes are necessary for LIPUS-induced promotion of fracture healing via transcriptional control of target genes, which presumably activates neighboring cells involved in fracture healing processes.
Collapse
|
18
|
Kong Q, Quan Y, Tian G, Zhou J, Liu X. Purinergic P2 Receptors: Novel Mediators of Mechanotransduction. Front Pharmacol 2021; 12:671809. [PMID: 34025431 PMCID: PMC8138185 DOI: 10.3389/fphar.2021.671809] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Accepted: 04/26/2021] [Indexed: 02/05/2023] Open
Abstract
Mechanosensing and mechanotransduction are vital processes in mechanobiology and play critical roles in regulating cellular behavior and fate. There is increasing evidence that purinergic P2 receptors, members of the purinergic family, play a crucial role in cellular mechanotransduction. Thus, information on the specific mechanism of P2 receptor-mediated mechanotransduction would be valuable. In this review, we focus on purinergic P2 receptor signaling pathways and describe in detail the interaction of P2 receptors with other mechanosensitive molecules, including transient receptor potential channels, integrins, caveolae-associated proteins and hemichannels. In addition, we review the activation of purinergic P2 receptors and the role of various P2 receptors in the regulation of various pathophysiological processes induced by mechanical stimuli.
Collapse
Affiliation(s)
- Qihang Kong
- Laboratory of Cardiovascular Diseases, Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, China
| | - Yue Quan
- Laboratory of Cardiovascular Diseases, Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, China
| | - Geer Tian
- Laboratory of Cardiovascular Diseases, Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, China
| | - Junteng Zhou
- Department of Cardiology, West China Hospital, Sichuan University, Chengdu, China
| | - Xiaojing Liu
- Laboratory of Cardiovascular Diseases, Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, China.,Department of Cardiology, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
19
|
Sun Y, Ge J, Tang W, Hong H, Liu D, Lin J. Hsa_circ_0045714 induced by eupatilin has a potential to promote fracture healing. Biofactors 2021; 47:376-385. [PMID: 33496034 DOI: 10.1002/biof.1707] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Accepted: 12/25/2020] [Indexed: 12/22/2022]
Abstract
It is thought that maintaining preosteoblast viability is constructive to fracture healing. Here, we explored the effects of eupatilin on preosteoblast and addressed the mechanism associated with hsa_circ_0045714. Blood specimens were collected from 32 patients with hand fracture or calcaneus fracture. MC3T3-E1 cells were treated with eupatilin. Small interfering-RNA was transfected into MC3T3-E1 cells. The ability of MC3T3-E1 cells to survive, proliferate, migrate, and express fracture-associated proteins was examined by 3-(4,5)-dimethylthiahiazo (-z-y1)-3,5-di- phenytetrazoliumromide (MTT), 5-bromodeoxyuridine (BrdU), 24-Transwell, Quantitative reverse transcription polymerase chain reaction (qRT-PCR), and Western blot. Hsa_circ_0045714 was detected by qRT-PCR. NF-κB and PI3K/AKT were evaluated by Western blot. Eupatilin enhanced the survival, proliferation, and migration of MC3T3-E1 cells. Cyclin D1, cyclin E, collagen II, aggrecan, and sulfated glycosaminoglycan (sGAG) were upregulated, while MMP-13 was downregulated in eupatilin-treated cells. Hsa_circ_0045714 was increased in patients with hand and calcaneus fractures with the time-lapse of surgical operation. In eupatilin-treated cells, Hsa_circ_0045714 was also elevated. However, the beneficial effects of eupatilin were weakened in hsa_circ_0045714-deficient cells. Molecularly, eupatilin-induced blockage of NF-κB and activation of PI3K/AKT were abrogated in hsa_circ_0045714-silenced cells. Our results confirmed the beneficial effects of eupatilin in preosteoblast, indicating eupatilin was a promising candidate for fracture healing.
Collapse
Affiliation(s)
- Yan Sun
- The First Ward of Trauma Orthopedics, Yantaishan Hospital, Yantai, Shandong, China
| | - Junbo Ge
- The First Ward of Trauma Orthopedics, Yantaishan Hospital, Yantai, Shandong, China
| | - Weiwei Tang
- Department of Tramatic Orthopedics, Yantaishan Hospital, Yantai, Shandong, China
| | - Huanyu Hong
- The First Ward of Trauma Orthopedics, Yantaishan Hospital, Yantai, Shandong, China
| | - Dong Liu
- Department of Orthopedics, Yantaishan Hospital, Yantai, Shandong, China
| | - Jiangtao Lin
- The First Ward of Trauma Orthopedics, Yantaishan Hospital, Yantai, Shandong, China
| |
Collapse
|
20
|
Janus 3D printed dynamic scaffolds for nanovibration-driven bone regeneration. Nat Commun 2021; 12:1031. [PMID: 33589620 PMCID: PMC7884435 DOI: 10.1038/s41467-021-21325-x] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Accepted: 01/13/2021] [Indexed: 12/12/2022] Open
Abstract
The application of physical stimuli to cell cultures has shown potential to modulate multiple cellular functions including migration, differentiation and survival. However, the relevance of these in vitro models to future potential extrapolation in vivo depends on whether stimuli can be applied “externally”, without invasive procedures. Here, we report on the fabrication and exploitation of dynamic additive-manufactured Janus scaffolds that are activated on-command via external application of ultrasounds, resulting in a mechanical nanovibration that is transmitted to the surrounding cells. Janus scaffolds were spontaneously formed via phase-segregation of biodegradable polycaprolactone (PCL) and polylactide (PLA) blends during the manufacturing process and behave as ultrasound transducers (acoustic to mechanical) where the PLA and PCL phases represent the active and backing materials, respectively. Remote stimulation of Janus scaffolds led to enhanced cell proliferation, matrix deposition and osteogenic differentiation of seeded human bone marrow derived stromal cells (hBMSCs) via formation and activation of voltage-gated calcium ion channels. Fabrication of dynamic, reversible and biocompatible scaffolds with non-invasive external triggers has so far been limited. Here, the authors report on the creation of 3D printed scaffolds with Janus structure that produce nanovibrations when exposed to ultrasound, promoting bone regeneration.
Collapse
|
21
|
Tang L, An S, Zhang Z, Fan X, Guo J, Sun L, Ta D. MSTN is a key mediator for low-intensity pulsed ultrasound preventing bone loss in hindlimb-suspended rats. Bone 2021; 143:115610. [PMID: 32829040 DOI: 10.1016/j.bone.2020.115610] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 08/17/2020] [Accepted: 08/18/2020] [Indexed: 12/13/2022]
Abstract
Low-intensity pulsed ultrasound (LIPUS) has been used to accelerate bone fracture healing. However, the issue whether LIPUS is effective in preventing osteoporosis has not been clarified, and if so, what possible mechanisms might be responsible. Myostatin (MSTN) is a negative regulator of muscle growth, and its absence will trigger a positive response to bone. In this study, we examined the effects of LIPUS on bone micro-structure, mechanical properties and damage healing of hindlimb-suspended rats, and investigated whether the inhibition of MSTN plays a role in this process. The rats were randomly divided into four groups: Normal control group (NC), Hind limb suspension group (HLS), Hind limb suspension and 80 mW/cm2 LIPUS irradiation group (HLS+ 80 mW/cm2), Hind limb suspension and 30 mW/cm2 LIPUS irradiation group (HLS+ 30 mW/cm2). The HLS+ 80 mW/cm2 rats were treated with LIPUS (1 MHz, 80 mW/cm2) and the HLS+ 30 mW/cm2 rats were treated with LIPUS (1 MHz, 30 mW/cm2) on the femur for 20 min/day for 28 days. MC3T3-E1 cells were respectively cultured with the serum of wild type mouse and MSTN knockout mouse at 1% concentration for 7 days. After 28 days, LIPUS effectively prevented the destruction of bone microstructure and the decline of mechanical properties, and promoted bone defect healing in the tail-suspended rats. In addition, LIPUS effectively reduced the MSTN content in the quadriceps and serum of the tail-suspended rats, inhibited its receptor and downstream signaling molecules and activated the Wnt signaling pathway in femurs. Growth of MC-3T3-E1 cell cultured with the serum of MSTN knockout mice was superior to that with wild mice serum on day 7. These results indicate that MSTN is a key mediator in LIPUS preventing bone loss caused by hindlimb-suspension.
Collapse
Affiliation(s)
- Liang Tang
- Institute of Sports Biology, Shaanxi Normal University, Xi'an 710119, China
| | - Shasha An
- Institute of Sports Biology, Shaanxi Normal University, Xi'an 710119, China
| | - Zhihao Zhang
- Institute of Sports Biology, Shaanxi Normal University, Xi'an 710119, China
| | - Xiushan Fan
- Institute of Sports Biology, Shaanxi Normal University, Xi'an 710119, China
| | - Jianzhong Guo
- Shaanxi Key Laboratory of Ultrasonics, Shaanxi Normal University, Xi'an 710119, China
| | - Lijun Sun
- Institute of Sports Biology, Shaanxi Normal University, Xi'an 710119, China.
| | - Dean Ta
- Department of Electronic Engineering, Fudan University, Shanghai 200433, China; Human Phenome Institute, Fudan University, Shanghai 201203, China; Key Laboratory of Medical Imaging Computing and Computer Assisted Intervention (MICCAI) of Shanghai, Shanghai 200032, China.
| |
Collapse
|
22
|
Jiang YX, Gong P, Zhang L. [A review of mechanisms by which low-intensity pulsed ultrasound affects bone regeneration]. HUA XI KOU QIANG YI XUE ZA ZHI = HUAXI KOUQIANG YIXUE ZAZHI = WEST CHINA JOURNAL OF STOMATOLOGY 2020; 38:571-575. [PMID: 33085244 DOI: 10.7518/hxkq.2020.05.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Low-intensity pulsed ultrasound (LIPUS) is a common physical therapy to accelerate the healing of bone fracture and treat delayed union of bone fracture. Vessels, nerves, and bone tissue are essential constituents of bone system. Recently, increasing evidence has been revealed that LIPUS can not only promote bone regeneration by directly regulating osteoblasts, osteoblasts, mesenchymal stem cells, but also have a positive impact on the repair of bone healing through vessels and nerves. Thus, we reviewed and summarized the latest published literature about the molecular mechanism for the effects of LIPUS on bone regeneration, which might offer a promising therapy for bone-related diseases.
Collapse
Affiliation(s)
- Yi-Xuan Jiang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Dept. of Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Ping Gong
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Dept. of Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Liang Zhang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Dept. of Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| |
Collapse
|
23
|
Inoue A, Nakao-Kuroishi K, Kometani-Gunjigake K, Mizuhara M, Shirakawa T, Ito-Sago M, Yasuda K, Nakatomi M, Matsubara T, Tada-Shigeyama Y, Morikawa K, Kokabu S, Kawamoto T. VNUT/SLC17A9, a vesicular nucleotide transporter, regulates osteoblast differentiation. FEBS Open Bio 2020; 10:1612-1623. [PMID: 32592329 PMCID: PMC7396442 DOI: 10.1002/2211-5463.12918] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 06/04/2020] [Accepted: 06/22/2020] [Indexed: 12/14/2022] Open
Abstract
Osteoblasts release adenosine triphosphate (ATP) out of the cell following mechanical stress. Although it is well established that extracellular ATP affects bone metabolism via P2 receptors [such as purinergic receptor P2X7 (P2X7R) and purinergic receptor P2Y2 (P2Y2R)], the mechanism of ATP release from osteoblasts remains unknown. Recently, a vesicular nucleotide transporter [VNUT, solute carrier family 17 member 9 (SLC17A9)] that preserves ATP in vesicles has been identified. The purpose of this study was to elucidate the role of VNUT in osteoblast bone metabolism. mRNA and protein expression of VNUT were confirmed in mouse bone and in osteoblasts by quantitative real-time PCR (qPCR) and immunohistochemistry. Next, when compressive force was applied to MC3T3-E1 cells by centrifugation, the expression of Slc17a9, P2x7r, and P2y2r was increased concomitant with an increase in extracellular ATP levels. Furthermore, compressive force decreased the osteoblast differentiation capacity of MC3T3-E1 cells. shRNA knockdown of Slc17a9 in MC3T3-E1 cells reduced levels of extracellular ATP and also led to increased osteoblast differentiation after the application of compressive force as assessed by qPCR analysis of osteoblast markers such as Runx2, Osterix, and alkaline phosphatase (ALP) as well as ALP activity. Consistent with these observations, knockdown of P2x7r or P2y2r by siRNA partially rescued the downregulation of osteoblast differentiation markers, caused by mechanical loading. In conclusion, our results demonstrate that VNUT is expressed in osteoblasts and that VNUT inhibits osteoblast differentiation in response to compressive force by mechanisms related to ATP release and P2X7R and/or P2Y2R activity.
Collapse
Affiliation(s)
- Asako Inoue
- Division of Orofacial Functions and Orthodontics, Department of Health Improvement, Kyushu Dental University, Kitakyushu-shi, Japan
| | - Kayoko Nakao-Kuroishi
- Division of Orofacial Functions and Orthodontics, Department of Health Improvement, Kyushu Dental University, Kitakyushu-shi, Japan
| | - Kaori Kometani-Gunjigake
- Division of Orofacial Functions and Orthodontics, Department of Health Improvement, Kyushu Dental University, Kitakyushu-shi, Japan
| | - Masahiro Mizuhara
- Division of Orofacial Functions and Orthodontics, Department of Health Improvement, Kyushu Dental University, Kitakyushu-shi, Japan
| | - Tomohiko Shirakawa
- Division of Orofacial Functions and Orthodontics, Department of Health Improvement, Kyushu Dental University, Kitakyushu-shi, Japan
| | - Misa Ito-Sago
- Division of Orofacial Functions and Orthodontics, Department of Health Improvement, Kyushu Dental University, Kitakyushu-shi, Japan
| | - Kazuma Yasuda
- Division of Orofacial Functions and Orthodontics, Department of Health Improvement, Kyushu Dental University, Kitakyushu-shi, Japan
| | - Mitsushiro Nakatomi
- Division of Anatomy, Department of Health Improvement, Kyushu Dental University, Kitakyushu-shi, Japan
| | - Takuma Matsubara
- Division of Molecular Signaling and Biochemistry, Department of Health Improvement, Kyushu Dental University, Kitakyushu-shi, Japan
| | - Yukiyo Tada-Shigeyama
- Division of Dental Anesthesiology, Department of Science of Physical Functions, Kyushu Dental University, Kitakyushu-shi, Japan
| | - Kazumasa Morikawa
- Division of Pediatric and Special Care Dentistry, Department of Developmental Oral Health Science, School of Dentistry, Iwate Medical University, Morioka, Japan
| | - Shoichiro Kokabu
- Division of Molecular Signaling and Biochemistry, Department of Health Improvement, Kyushu Dental University, Kitakyushu-shi, Japan
| | - Tatsuo Kawamoto
- Division of Orofacial Functions and Orthodontics, Department of Health Improvement, Kyushu Dental University, Kitakyushu-shi, Japan
| |
Collapse
|
24
|
Rahimi A, Case N. Computational model to evaluate modulation of the acoustic field in an ultrasonic bioreactor by incorporation of a water layer bounded by an acoustic absorbent boundary layer. ULTRASONICS 2020; 103:106086. [PMID: 32070827 DOI: 10.1016/j.ultras.2020.106086] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Revised: 12/04/2019] [Accepted: 12/06/2019] [Indexed: 06/10/2023]
Abstract
Ultrasonic bioreactors have been used for in vitro experimentation to study cellular responses to low-intensity pulsed ultrasound. The presence of an air interface in these bioreactors contributes to variability in the acoustic pressure field, reducing experimental reproducibility. A multiphysics finite element model was developed to simulate the acoustic field in an in-dish ultrasonic bioreactor, where the transducer is immersed in culture medium above the dish surface, and the effects of replacing air below the dish in the bioreactor with a water layer bounded by an acoustic absorbent layer were evaluated. Frequency domain simulations showed that the spatially-averaged pressure at the dish surface alternated between a minimum and maximum level as the distance between the dish and transducer increased. The ratio of the maximum to minimum level was 6.5-fold when the air interface was present, and this ratio dropped to 1.8-fold with replacement of the air interface. However, radial pressure variability was present with or without the air interface in the bioreactor model. Time-dependent simulations showed that the increase in acoustic pressure to a maximum level after US signal activation and the pressure drop after signal cessation were faster when the water-coupled non-reflective layer was used to replace the air layer below the dish, generating a pressure pattern that more closely followed the applied pulsed ultrasound signal due to reduced wave reflection and interference. Overall, this work showed that having water rather than air in contact with the lower dish surface when paired with an acoustic absorbent layer resulted in a less variable pressure field, providing an improved bioreactor design for in vitro experiments.
Collapse
Affiliation(s)
- Abdolrasol Rahimi
- Biomedical Engineering, Parks College of Engineering, Aviation and Technology, Saint Louis University, Saint Louis, MO 63103, USA
| | - Natasha Case
- Biomedical Engineering, Parks College of Engineering, Aviation and Technology, Saint Louis University, Saint Louis, MO 63103, USA.
| |
Collapse
|
25
|
Guan M, Zhu Y, Liao B, Tan Q, Qi H, Zhang B, Huang J, Du X, Bai D. Low-intensity pulsed ultrasound inhibits VEGFA expression in chondrocytes and protects against cartilage degeneration in experimental osteoarthritis. FEBS Open Bio 2020; 10:434-443. [PMID: 31975545 PMCID: PMC7050266 DOI: 10.1002/2211-5463.12801] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Revised: 01/09/2020] [Accepted: 01/21/2020] [Indexed: 12/16/2022] Open
Abstract
Low-intensity pulsed ultrasound (LIPUS), a noninvasive physical therapy, was recently demonstrated to be an effective treatment for osteoarthritis (OA). Vascular endothelium growth factor A (VEGFA) has been found to be upregulated in the articular cartilage, synovium and subchondral bone of OA patients, leading to cartilage degeneration, synovitis and osteophyte formation. However, the functions and mechanisms of LIPUS in regulating chondrocyte-derived VEGFA expression are still unclear. In this study, we investigated whether LIPUS attenuated OA progression by (a) decreasing the percentage of VEGFA-positive cells in mouse articular cartilage destabilised through medial meniscus surgery and (b) relieving interleukin-1β-induced VEGFA expression in mouse primary chondrocytes. However, this function was negated by a p38 mitogen-activated protein kinase (p38 MAPK) inhibitor. In addition, we found that LIPUS ameliorated VEGFA-mediated disorders in cartilage extracellular matrix metabolism and chondrocyte hypertrophy during OA development. In conclusion, our data indicate a novel effect of LIPUS in regulating the expression of osteoarthritic chondrocyte-derived VEGFA through the suppression of p38 MAPK activity.
Collapse
Affiliation(s)
- Mengtong Guan
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Chongqing Medical University, China
| | - Ying Zhu
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Chongqing Medical University, China
| | - Bo Liao
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Chongqing Medical University, China
| | - Qiaoyan Tan
- Department of Rehabilitation Medicine, Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Huabing Qi
- Department of Rehabilitation Medicine, Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Bin Zhang
- Department of Rehabilitation Medicine, Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Junlan Huang
- Department of Rehabilitation Medicine, Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Xiaolan Du
- Department of Rehabilitation Medicine, Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Dingqun Bai
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Chongqing Medical University, China
| |
Collapse
|
26
|
Corciulo C, Cronstein BN. Signaling of the Purinergic System in the Joint. Front Pharmacol 2020; 10:1591. [PMID: 32038258 PMCID: PMC6993121 DOI: 10.3389/fphar.2019.01591] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Accepted: 12/09/2019] [Indexed: 12/15/2022] Open
Abstract
The joint is a complex anatomical structure consisting of different tissues, each with a particular feature, playing together to give mobility and stability at the body. All the joints have a similar composition including cartilage for reducing the friction of the movement and protecting the underlying bone, a synovial membrane that produces synovial fluid to lubricate the joint, ligaments to limit joint movement, and tendons for the interaction with muscles. Direct or indirect damage of one or more of the tissues forming the joint is the foundation of different pathological conditions. Many molecular mechanisms are involved in maintaining the joint homeostasis as well as in triggering disease development. The molecular pathway activated by the purinergic system is one of them.The purinergic signaling defines a group of receptors and intermembrane channels activated by adenosine, adenosine diphosphate, adenosine 5’-triphosphate, uridine triphosphate, and uridine diphosphate. It has been largely described as a modulator of many physiological and pathological conditions including rheumatic diseases. Here we will give an overview of the purinergic system in the joint describing its expression and function in the synovium, cartilage, ligament, tendon, and bone with a therapeutic perspective.
Collapse
Affiliation(s)
- Carmen Corciulo
- Division of Translational Medicine, Department of Medicine, NYU School of Medicine, New York, NY, United States.,Krefting Research Centre-Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Bruce N Cronstein
- Division of Translational Medicine, Department of Medicine, NYU School of Medicine, New York, NY, United States.,Division of Rheumatology, Department of Medicine, NYU School of Medicine, New York, NY, United States
| |
Collapse
|
27
|
Sun S, Sun L, Kang Y, Tang L, Qin YX, Ta D. Therapeutic Effects of Low-Intensity Pulsed Ultrasound on Osteoporosis in Ovariectomized Rats: Intensity-Dependent Study. ULTRASOUND IN MEDICINE & BIOLOGY 2020; 46:108-121. [PMID: 31587953 DOI: 10.1016/j.ultrasmedbio.2019.08.025] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 08/13/2019] [Accepted: 08/30/2019] [Indexed: 06/10/2023]
Abstract
This study investigated the effects of low-intensity pulsed ultrasound (LIPUS) of different spatial-average-temporal-average intensity (ISATA) ranging from 15-150 mW/cm2 on the treatment of osteoporosis in ovariectomized rats. Healthy 3-mo-old female Sprague-Dawley rats were randomly divided into nine groups (n = 12 per group): sham-ovariectomy (OVX) control group, OVX control group and OVX groups treated with LIPUS at seven different intensities (ISATA: 15, 30, 50, 75, 100, 125 and 150 mW/cm2, respectively). LIPUS was applied to bilateral femurs 12 wk post-OVX for 20 min/d for 6 wk. Micro-computed tomography, biomechanical tests, serum biochemical analysis and grip strength tests were performed to evaluate the therapeutic effects of LIPUS at different intensities. Results revealed that LIPUS intensity yielded strong correlations with bone mineral density and bone microstructure (R2 = 0.57-0.83) and bone mechanical strength (R2 = 0.80-0.97), and that the intensity of 150 mW/cm2, instead of the 30 mW/cm2 widely used in bone fracture healing, was most effective in maintaining bone mass among all the LIPUS signals between 15 and 150 mW/cm2. This suggests that higher ultrasound intensity (i.e., 150 mW/cm2) may be more effective than lower intensity in mitigation of osteopenia and osteoporosis.
Collapse
Affiliation(s)
- Shuxin Sun
- Department of Electronic Engineering, Fudan University, Shanghai, China
| | - Lijun Sun
- Institute of Sports Biology, Shaanxi Normal University, Xi'an, China
| | - Yiting Kang
- Institute of Sports Biology, Shaanxi Normal University, Xi'an, China
| | - Liang Tang
- Institute of Sports Biology, Shaanxi Normal University, Xi'an, China
| | - Yi-Xian Qin
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, New York, USA
| | - Dean Ta
- Department of Electronic Engineering, Fudan University, Shanghai, China; State Key Laboratory of ASIC and System, Fudan University, Shanghai, China; Key Laboratory of Medical Imaging Computing and Computer Assisted Intervention (MICCAI) of Shanghai, Shanghai, China.
| |
Collapse
|
28
|
Kusuyama J, Seong C, Makarewicz NS, Ohnishi T, Shima K, Semba I, Bandow K, Matsuguchi T. Low intensity pulsed ultrasound (LIPUS) maintains osteogenic potency by the increased expression and stability of Nanog through spleen tyrosine kinase (Syk) activation. Cell Signal 2019; 62:109345. [PMID: 31228531 DOI: 10.1016/j.cellsig.2019.109345] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Revised: 05/22/2019] [Accepted: 06/18/2019] [Indexed: 12/18/2022]
Abstract
Mesenchymal stem cells (MSCs) are a powerful tool for cell-based, clinical therapies like bone regeneration. Therapeutic use of cell transplantation requires many cells, however, the expansion process needed to produce large quantities of cells reduces the differentiation potential of MSCs. Here, we examined the protective effects of low intensity pulsed ultrasound (LIPUS) on the maintenance of osteogenic potency. Primary osteoblastic cells were serially passaged between 2 and 12 times with daily LIPUS treatment. We found that LIPUS stimulation maintains osteogenic differentiation capacity in serially passaged cells, as characterized by improved matrix mineralization and Osteocalcin mRNA expression. Decreased expression of Nanog, Sox2, and Msx2, and increased expression of Pparg2 from serial passaging was recovered in LIPUS-stimulated cells. We found that LIPUS stimulation not only increased but also sustained expression of Nanog in primary osteoblasts and ST2 cells, a mouse mesenchymal stromal cell line. Nanog overexpression in serially passaged cells mimicked the recuperative effects of LIPUS on osteogenic potency, highlighting the important role of Nanog in LIPUS stimulation. Additionally, we found that spleen tyrosine kinase (Syk) is an important signaling molecule to induce Nanog expression in LIPUS-stimulated cells. Syk activation was regulated by both Rho-associated kinase 1 (ROCK1) and extracellular ATP in a paracrine manner. Interestingly, the LIPUS-induced increase in Nanog mRNA expression was regulated by ATP-P2X4-Syk Y323 activation, while the improvement of Nanog protein stability was controlled by the ROCK1-Syk Y525/526 pathway. Taken together, these results indicate that LIPUS stimulation recovers and maintains the osteogenic potency of serially passaged cells through a Syk-Nanog axis.
Collapse
Affiliation(s)
- Joji Kusuyama
- Department of Oral Biochemistry, Field of Developmental Medicine, Kagoshima University Graduate School of Medical and Dental Sciences, 8-35-1 Sakuragaoka, Kagoshima 890-8544, Japan; Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Department of Medicine, Harvard Medical School, One Joslin Place, Boston, MA 02215, USA.
| | - Changhwan Seong
- Department of Oral Biochemistry, Field of Developmental Medicine, Kagoshima University Graduate School of Medical and Dental Sciences, 8-35-1 Sakuragaoka, Kagoshima 890-8544, Japan; Department of Oral and Maxillofacial Surgery, Field of Oral and Maxillofacial Rehabilitation, Kagoshima University Graduate School of Medical and Dental Sciences, 8-35-1 Sakuragaoka, Kagoshima 890-8544, Japan
| | - Nathan S Makarewicz
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Department of Medicine, Harvard Medical School, One Joslin Place, Boston, MA 02215, USA
| | - Tomokazu Ohnishi
- Department of Oral Biochemistry, Field of Developmental Medicine, Kagoshima University Graduate School of Medical and Dental Sciences, 8-35-1 Sakuragaoka, Kagoshima 890-8544, Japan
| | - Kaori Shima
- Department of Oral Pathology, Field of Oncology, Kagoshima University Graduate School of Medical and Dental Sciences, 8-35-1 Sakuragaoka, Kagoshima 890-8544, Japan
| | - Ichiro Semba
- Department of Oral Pathology, Field of Oncology, Kagoshima University Graduate School of Medical and Dental Sciences, 8-35-1 Sakuragaoka, Kagoshima 890-8544, Japan
| | - Kenjiro Bandow
- Department of Oral Biology and Tissue Engineering, Meikai University School of Dentistry, 1-1 Keyakidai, Sakato 350-0283, Saitama, Japan
| | - Tetsuya Matsuguchi
- Department of Oral Biochemistry, Field of Developmental Medicine, Kagoshima University Graduate School of Medical and Dental Sciences, 8-35-1 Sakuragaoka, Kagoshima 890-8544, Japan
| |
Collapse
|
29
|
Ottensmeyer PF, Witzler M, Schulze M, Tobiasch E. Small Molecules Enhance Scaffold-Based Bone Grafts via Purinergic Receptor Signaling in Stem Cells. Int J Mol Sci 2018; 19:E3601. [PMID: 30441872 PMCID: PMC6274752 DOI: 10.3390/ijms19113601] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Revised: 11/08/2018] [Accepted: 11/09/2018] [Indexed: 12/15/2022] Open
Abstract
The need for bone grafts is high, due to age-related diseases, such as tumor resections, but also accidents, risky sports, and military conflicts. The gold standard for bone grafting is the use of autografts from the iliac crest, but the limited amount of accessible material demands new sources of bone replacement. The use of mesenchymal stem cells or their descendant cells, namely osteoblast, the bone-building cells and endothelial cells for angiogenesis, combined with artificial scaffolds, is a new approach. Mesenchymal stem cells (MSCs) can be obtained from the patient themselves, or from donors, as they barely cause an immune response in the recipient. However, MSCs never fully differentiate in vitro which might lead to unwanted effects in vivo. Interestingly, purinergic receptors can positively influence the differentiation of both osteoblasts and endothelial cells, using specific artificial ligands. An overview is given on purinergic receptor signaling in the most-needed cell types involved in bone metabolism-namely osteoblasts, osteoclasts, and endothelial cells. Furthermore, different types of scaffolds and their production methods will be elucidated. Finally, recent patents on scaffold materials, as wells as purinergic receptor-influencing molecules which might impact bone grafting, are discussed.
Collapse
Affiliation(s)
- Patrick Frank Ottensmeyer
- Department of Natural Sciences, Bonn-Rhine-Sieg University of Applied Sciences, D-53359 Rheinbach, Germany.
| | - Markus Witzler
- Department of Natural Sciences, Bonn-Rhine-Sieg University of Applied Sciences, D-53359 Rheinbach, Germany.
| | - Margit Schulze
- Department of Natural Sciences, Bonn-Rhine-Sieg University of Applied Sciences, D-53359 Rheinbach, Germany.
| | - Edda Tobiasch
- Department of Natural Sciences, Bonn-Rhine-Sieg University of Applied Sciences, D-53359 Rheinbach, Germany.
| |
Collapse
|
30
|
Sekino J, Nagao M, Kato S, Sakai M, Abe K, Nakayama E, Sato M, Nagashima Y, Hino H, Tanabe N, Kawato T, Maeno M, Suzuki N, Ueda K. Low-intensity pulsed ultrasound induces cartilage matrix synthesis and reduced MMP13 expression in chondrocytes. Biochem Biophys Res Commun 2018; 506:290-297. [DOI: 10.1016/j.bbrc.2018.10.063] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Accepted: 10/10/2018] [Indexed: 01/12/2023]
|
31
|
Mikolajewicz N, Zimmermann EA, Willie BM, Komarova SV. Mechanically stimulated ATP release from murine bone cells is regulated by a balance of injury and repair. eLife 2018; 7:37812. [PMID: 30324907 PMCID: PMC6205812 DOI: 10.7554/elife.37812] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Accepted: 09/28/2018] [Indexed: 02/06/2023] Open
Abstract
Bone cells sense and actively adapt to physical perturbations to prevent critical damage. ATP release is among the earliest cellular responses to mechanical stimulation. Mechanical stimulation of a single murine osteoblast led to the release of 70 ± 24 amole ATP, which stimulated calcium responses in neighboring cells. Osteoblasts contained ATP-rich vesicles that were released upon mechanical stimulation. Surprisingly, interventions that promoted vesicular release reduced ATP release, while inhibitors of vesicular release potentiated ATP release. Searching for an alternative ATP release route, we found that mechanical stresses induced reversible cell membrane injury in vitro and in vivo. Ca2+/PLC/PKC-dependent vesicular exocytosis facilitated membrane repair, thereby minimizing cell injury and reducing ATP release. Priming cellular repair machinery prior to mechanical stimulation reduced subsequent membrane injury and ATP release, linking cellular mechanosensitivity to prior mechanical exposure. Thus, our findings position ATP release as an integrated readout of membrane injury and repair. Athletes' skeletons get stronger with training, while bones weaken in people who cannot move or in astronauts experiencing weightlessness. This is because bone cells thrive when exposed to forces. When a bone cell is exposed to a physical force, the first thing that happens is the release of the energy-rich molecule called ATP into the space outside the cell. This molecule then binds to the neighboring cell to unleash a cascade of responses. ATP can exit the cell either through special canals in the cell membrane or released in tiny pod-like structures called vesicles. It is known that strong forces can injure the cell membrane and cause ATP to spill out. However, it is less clear how ATP is released when cells are subjected to regular forces. Mikolajewicz et al. investigated whether ATP exits through injured membranes of cells experiencing regular forces. Bone cells grown in the laboratory were gently poked with a glass needle or placed in a turbulent fluid to simulate forces experienced in the body. Dyes and fluorescent imaging techniques were used to observe the movement of vesicles and calculate the concentration of ATP in these cells. The experiments showed that regular forces in the body do indeed injure the cell membranes and cause ATP to spill out. But importantly, the cells repaired the injuries quickly by releasing vesicles that patch the wound. As soon as the membrane is sealed, ATP stops coming out. From the first injury, cells adapted and quickly strengthened their membrane and repair system to be more resilient against future forces. This process was also seen in the shin bones of mice. These results are important because knowing how bone cells sense, respond and convert physical forces can help us develop treatments for astronauts, the injured and aged.
Collapse
Affiliation(s)
- Nicholas Mikolajewicz
- Faculty of Dentistry, McGill University, Montreal, Quebec, Canada.,Shriners Hospital for Children - Canada, Montreal, Quebec, Canada
| | - Elizabeth A Zimmermann
- Shriners Hospital for Children - Canada, Montreal, Quebec, Canada.,Department of Pediatric Surgery, Montreal, Quebec, Canada
| | - Bettina M Willie
- Shriners Hospital for Children - Canada, Montreal, Quebec, Canada.,Department of Pediatric Surgery, Montreal, Quebec, Canada
| | - Svetlana V Komarova
- Faculty of Dentistry, McGill University, Montreal, Quebec, Canada.,Shriners Hospital for Children - Canada, Montreal, Quebec, Canada
| |
Collapse
|
32
|
Zeng D, Yao P, Zhao H. P2X7, a critical regulator and potential target for bone and joint diseases. J Cell Physiol 2018; 234:2095-2103. [PMID: 30317598 DOI: 10.1002/jcp.27544] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2018] [Accepted: 09/13/2018] [Indexed: 12/18/2022]
Abstract
Abundant evidence indicted that P2X7 receptor show a essential role in human health and some human diseases including hypertension, atherosclerosis, pulmonary inflammation, tuberculosis infection, psychiatric disorders, and cancer. P2X7 receptor also has an important role in some central nervous system diseases such as neurodegenerative disorders. Recently, more research suggested that P2X7 receptor also plays a crucial role in bone and joint diseases. But the effect of P2X7 receptor on skeletal and joint diseases has not been systematically reviewed. In this article, the role of P2X7 receptor in skeletal and joint diseases is elaborated. The activation of P2X7 receptor can ameliorate osteoporosis by inducing a fine balance between osteoclastic resorption and osteoblastic bone formation. The activation of P2X7 receptor can relieve the stress fracture injury by increasing the response to mechanical loading and inducing osteogenesis. But the activation of P2X7 receptor mediates the cell growth and cell proliferation in bone cancer. In addition, the activation of P2X7 receptor can aggravate the process of some joint diseases such as osteoarthritis, rheumatoid arthritis, and acute gouty arthritis. The inhibition of P2X7 receptor can alleviate the pathological process of joint disease to some extent. In conclusion, P2X7 receptor may be a critical regulator and therapeutic target for bone and joint diseases.
Collapse
Affiliation(s)
- Dehui Zeng
- Department of Orthopedics, Affiliated Nanhua Hospital, University of South China, Hengyang, China
| | - Pingbo Yao
- Department of Orthopedics, Affiliated Nanhua Hospital, University of South China, Hengyang, China
| | - Hong Zhao
- Institute of Pharmacy and Pharmacology, Nursing College, University of South China, Hengyang, China
| |
Collapse
|
33
|
Wu S, Xu X, Sun J, Zhang Y, Shi J, Xu T. Low-Intensity Pulsed Ultrasound Accelerates Traumatic Vertebral Fracture Healing by Coupling Proliferation of Type H Microvessels. JOURNAL OF ULTRASOUND IN MEDICINE : OFFICIAL JOURNAL OF THE AMERICAN INSTITUTE OF ULTRASOUND IN MEDICINE 2018; 37:1733-1742. [PMID: 29363151 DOI: 10.1002/jum.14525] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Revised: 09/04/2017] [Accepted: 10/08/2017] [Indexed: 06/07/2023]
Abstract
OBJECTIVES Patients with traumatic vertebral fractures often have major associated postoperative morbidities such as healing failure and kyphosis. Low-intensity pulsed ultrasound (US) has been found to promote bone fracture healing. The objectives of our study were to determine whether low-intensity pulsed US could promote traumatic vertebral fracture healing and to explore its inner mechanisms. METHODS A rat model of traumatic vertebral fracture was created and treated with low-intensity pulsed US after surgery. At 4 weeks after surgery, radiographic, micro-computed tomography, and 3-dimensional reconstruction were used to assess the radiologic healing status; a histologic analysis was performed to evaluate the pathologic process and relationship between osteogenesis and type H microvessels. RESULTS Well-remodeled trabecular meshworks were found in the low-intensity pulsed US treatment group compared to the control group. Micro-computed tomography and 3-dimensional reconstruction revealed more and thicker trabeculae after low-intensity pulsed US treatment. Abundant chondrocytes, a newly formed bone marrow cavity, trabeculae, and microvessels were formed at the fracture sites. More osterix-positive osteoblasts were circling the newly formed bone meshwork and were situated at the interface of chondrocytes in the low-intensity pulsed US treatment group. Type H microvessels were spreading around the newly formed trabecula, bone marrow cavity, osteoblasts, and interface of chondrocytes, with a larger mean vascular density in the low-intensity pulsed US group. CONCLUSIONS Low-intensity pulsed US could accelerate traumatic vertebral fracture healing by temporally and spatially increasing chondrogenesis and osteoblast-induced osteogenesis coupled with angiogenesis of type H microvessels in a rat model of traumatic vertebral fracture.
Collapse
Affiliation(s)
- Suiyi Wu
- Faculty of Naval Medicine, Second Military Medical University, Shanghai, China
| | - Ximing Xu
- Department of Spine Surgery, Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Jingchuan Sun
- Department of Spine Surgery, Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Yao Zhang
- Cadet Brigade, Second Military Medical University, Shanghai, China
| | - Jiangang Shi
- Department of Spine Surgery, Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Tianming Xu
- 455 Hospital of Chinese People's Liberation Army, Shanghai, China
| |
Collapse
|
34
|
Matsuike R, Tanaka H, Nakai K, Kanda M, Nagasaki M, Murakami F, Shibata C, Mayahara K, Nakajima A, Tanabe N, Kawato T, Maeno M, Shimizu N. Continuous application of compressive force induces fusion of osteoclast-like RAW264.7 cells via upregulation of RANK and downregulation of LGR4. Life Sci 2018; 201:30-36. [PMID: 29572179 DOI: 10.1016/j.lfs.2018.03.038] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Revised: 03/12/2018] [Accepted: 03/19/2018] [Indexed: 01/08/2023]
Abstract
AIMS During orthodontic treatment, facilitating osteoclastic bone resorption in the alveolar bone exposed to the compressive force (CF) is an important factor for tooth movement. The present study investigated the effect of CF stimulation on the differentiation of RAW264.7 cells from precursors to mature osteoclasts. MAIN METHODS The cells were continuously stimulated with 0.3, 0.6, or 1.1 g/cm2 CF-which was generated by increasing the volume of culture medium in the wells of a 96-well plate-in the presence or absence of receptor activator of nuclear factor κB (RANK) ligand (RANKL) for 4 days. KEY FINDINGS In the presence of RANKL, the number of tartrate-resistant acid phosphatase (TRAP)-positive multinucleated cells and the mRNA levels of dendritic cell-specific transmembrane protein (DC-STAMP) and osteoclast-stimulatory transmembrane protein (OC-STAMP) were increased by application of 0.6 and 1.1 g/cm2 CF as compared to 0.3 g/cm2 CF. The mRNA level of RANK was upregulated whereas that of leucine-rich repeat-containing G-protein-coupled receptor (LGR)4-another RANKL receptor was downregulated by 0.6 and 1.1 g/cm2 CF as compared to 0.3 g/cm2 CF in the absence of RANKL. The proportion of cells with nuclear translocation of the nuclear translocation of nuclear factor of activated T cells (NFAT)c1 was increased by 0.6 and 1.1 g/cm2 CF in the presence of RANKL. SIGNIFICANCE Continuous application of CF induced the differentiation of RAW264.7 cells into TRAP-positive multinuclear cells by enhancing the expression of DC- and OC-STAMP and the nuclear translocation of NFATc1. This may result from the CF-induced increase in RANK and decrease in LGR4 expression.
Collapse
Affiliation(s)
- Rieko Matsuike
- Nihon University Graduate School of Dentistry, Tokyo, Japan
| | - Hideki Tanaka
- Department of Oral Health Sciences, Nihon University School of Dentistry, Tokyo, Japan; Division of Functional Morphology, Dental Research Center, Nihon University School of Dentistry, Tokyo, Japan
| | - Kumiko Nakai
- Department of Oral Health Sciences, Nihon University School of Dentistry, Tokyo, Japan; Division of Functional Morphology, Dental Research Center, Nihon University School of Dentistry, Tokyo, Japan
| | - Mai Kanda
- Nihon University Graduate School of Dentistry, Tokyo, Japan
| | - Maki Nagasaki
- Nihon University Graduate School of Dentistry, Tokyo, Japan
| | | | - Chika Shibata
- Nihon University Graduate School of Dentistry, Tokyo, Japan
| | - Kotoe Mayahara
- Department of Orthodontics, Nihon University School of Dentistry, Tokyo, Japan; Division of Clinical Research, Dental Research Center, Nihon University School of Dentistry, Tokyo, Japan
| | - Akira Nakajima
- Department of Orthodontics, Nihon University School of Dentistry, Tokyo, Japan; Division of Clinical Research, Dental Research Center, Nihon University School of Dentistry, Tokyo, Japan
| | - Natsuko Tanabe
- Division of Functional Morphology, Dental Research Center, Nihon University School of Dentistry, Tokyo, Japan; Department of Biochemistry, Nihon University School of Dentistry, Tokyo, Japan
| | - Takayuki Kawato
- Department of Oral Health Sciences, Nihon University School of Dentistry, Tokyo, Japan; Division of Functional Morphology, Dental Research Center, Nihon University School of Dentistry, Tokyo, Japan.
| | - Masao Maeno
- Department of Oral Health Sciences, Nihon University School of Dentistry, Tokyo, Japan; Division of Functional Morphology, Dental Research Center, Nihon University School of Dentistry, Tokyo, Japan
| | - Noriyoshi Shimizu
- Department of Orthodontics, Nihon University School of Dentistry, Tokyo, Japan; Division of Clinical Research, Dental Research Center, Nihon University School of Dentistry, Tokyo, Japan
| |
Collapse
|
35
|
Tassinary JAF, Lunardelli A, Basso BDS, Dias HB, Catarina AV, Stülp S, Haute GV, Martha BA, Melo DADS, Nunes FB, Donadio MVF, Oliveira JRD. Low-intensity pulsed ultrasound (LIPUS) stimulates mineralization of MC3T3-E1 cells through calcium and phosphate uptake. ULTRASONICS 2018; 84:290-295. [PMID: 29182945 DOI: 10.1016/j.ultras.2017.11.011] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Revised: 11/20/2017] [Accepted: 11/20/2017] [Indexed: 06/07/2023]
Abstract
The present study aimed to evaluate the effect of low-intensity pulsed ultrasound (LIPUS) on pre-osteoblast mineralization using in vitro bioassays. Pre-osteoblastic MC3T3-E1 cells were exposed to LIPUS at 1 MHz frequency, 0.2 W/cm2 intensity and 20% duty cycle for 30 min. The analyses were carried out up to 336 h (14 days) after exposure. The concentration of collagen, phosphate, alkaline phosphatase, calcium and transforming growth factor beta 1 (TGF-β1) in cell supernatant and the presence of calcium deposits in the cells were analyzed. Our results showed that LIPUS promotes mineralized nodules formation. Collagen, phosphate, and calcium levels were decreased in cell supernatant at 192 h after LIPUS exposure. However, alkaline phosphatase and TGF-β1 concentrations remained unchanged. Therapeutic pulsed ultrasound is capable of stimulating differentiation and mineralization of pre-osteoblastic MC3T3-E1 cells by calcium and phosphate uptake with consequent hydroxyapatite formation.
Collapse
Affiliation(s)
- João Alberto Fioravante Tassinary
- Univates, Lajeado, Rio Grande do Sul, Brazil; Laboratório de Pesquisa em Biofísica Celular e Inflamação, Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, Rio Grande do Sul, Brazil
| | - Adroaldo Lunardelli
- Laboratório de Pesquisa em Biofísica Celular e Inflamação, Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, Rio Grande do Sul, Brazil; Centro Universitário Ritter dos Reis (UniRitter), Porto Alegre, Rio Grande do Sul, Brazil
| | - Bruno de Souza Basso
- Laboratório de Pesquisa em Biofísica Celular e Inflamação, Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, Rio Grande do Sul, Brazil
| | - Henrique Bregolin Dias
- Laboratório de Pesquisa em Biofísica Celular e Inflamação, Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, Rio Grande do Sul, Brazil
| | - Anderson Velasque Catarina
- Laboratório de Pesquisa em Biofísica Celular e Inflamação, Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, Rio Grande do Sul, Brazil
| | | | - Gabriela Viegas Haute
- Laboratório de Pesquisa em Biofísica Celular e Inflamação, Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, Rio Grande do Sul, Brazil
| | - Bianca Andrade Martha
- Laboratório de Pesquisa em Biofísica Celular e Inflamação, Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, Rio Grande do Sul, Brazil
| | - Denizar Alberto da Silva Melo
- Laboratório de Pesquisa em Biofísica Celular e Inflamação, Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, Rio Grande do Sul, Brazil
| | - Fernanda Bordignon Nunes
- Laboratório de Pesquisa em Biofísica Celular e Inflamação, Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, Rio Grande do Sul, Brazil
| | - Márcio Vinícius Fagundes Donadio
- Laboratório de Pesquisa em Biofísica Celular e Inflamação, Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, Rio Grande do Sul, Brazil
| | - Jarbas Rodrigues de Oliveira
- Laboratório de Pesquisa em Biofísica Celular e Inflamação, Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, Rio Grande do Sul, Brazil.
| |
Collapse
|
36
|
PYK2 mediates BzATP-induced extracellular matrix proteins synthesis. Biochem Biophys Res Commun 2017; 494:663-667. [PMID: 29061307 DOI: 10.1016/j.bbrc.2017.10.107] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2017] [Accepted: 10/20/2017] [Indexed: 10/25/2022]
Abstract
Mechanical stimuli such as fluid shear and cyclic tension force induced extracellular adenosine triphosphate (ATP) release in osteoblasts. In particular, cyclic tension force-induced ATP enhances bone formation through P2X7 activation. Proline-rich tyrosine kinase 2 (PYK2) mediate osteoblasts differentiation is induced by mechanical stimuli. Furthermore, activation of PYK2 also was a response to integrin by mechanical stimuli. Extracellular matrix protein (ECMP)s, which are important factors for bone formation are expressed by osteoblasts. However, the effect of the interaction of 2'(3)-Ο-(4-Benzoylbenzoyl) adenosine-5'-triphosphate (BzATP), which is the agonist of the mechanosensitive receptor P2X7, with PYK2 on ECMP production is poorly understood. Thus, our purpose was to investigate the effects of PYK2 on BzATP-induced ECMP production in osteoblasts. BzATP increased phospho-PYK2 protein expression on days 3 and 7 of culture. Furthermore, the PYK2 inhibitor PF431394 inhibited the stimulatory effect of BzATP on the expression of type I collagen, bone sialoprotein and osteocalcin expression. PF431396 did not inhibit the stimulatory effect of BzATP on osteopontin (OPN) mRNA expression. These results suggest that mechanical stimuli activate P2X7 might induce ECMPs expression through PYK2 except in the case of OPN expression. Altogether, mechanical stimuli-induced ECMPs production might be implicated by extracellular ATP secretion or integrin via PYK2 activation.
Collapse
|
37
|
Minashima T, Quirno M, Lee YJ, Kirsch T. The role of the progressive ankylosis protein (ANK) in adipogenic/osteogenic fate decision of precursor cells. Bone 2017; 98:38-46. [PMID: 28286238 PMCID: PMC5396059 DOI: 10.1016/j.bone.2017.03.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Revised: 03/06/2017] [Accepted: 03/07/2017] [Indexed: 01/14/2023]
Abstract
The progressive ankylosis protein (ANK) is a transmembrane protein that transports intracellular pyrophosphate (PPi) to the extracellular milieu. In this study we show increased fatty degeneration of the bone marrow of adult ank/ank mice, which lack a functional ANK protein. In addition, isolated bone marrow stromal cells (BMSCs) isolated from ank/ank mice showed a decreased proliferation rate and osteogenic differentiation potential, and an increased adipogenic differentiation potential compared to BMSCs isolated from wild type (WT) littermates. Wnt signaling pathway PCR array analysis revealed that Wnt ligands, Wnt receptors and Wnt signaling proteins that stimulate osteoblast differentiation were expressed at markedly lower levels in ank/ank BMSCs than in WT BMSCs. Lack of ANK function also resulted in impaired bone fracture healing, as indicated by a smaller callus formed and delayed bone formation in the callus site. Whereas 5weeks after fracture, the fractured bone in WT mice was further remodeled and restored to original shape, the fractured bone in ank/ank mice was not fully restored and remodeled to original shape. In conclusion, our study provides evidence that ANK plays a critical role in the adipogenic/osteogenic fate decision of adult mesenchymal precursor cells. ANK functions in precursor cells are required for osteogenic differentiation of these cells during adult bone homeostasis and repair, whereas lack of ANK functions favors adipogenic differentiation.
Collapse
Affiliation(s)
- Takeshi Minashima
- Musculoskeletal Research Center, Department of Orthopaedic Surgery, New York University School of Medicine, NY, New York, United States
| | - Martin Quirno
- Musculoskeletal Research Center, Department of Orthopaedic Surgery, New York University School of Medicine, NY, New York, United States
| | - You Jin Lee
- Musculoskeletal Research Center, Department of Orthopaedic Surgery, New York University School of Medicine, NY, New York, United States
| | - Thorsten Kirsch
- Musculoskeletal Research Center, Department of Orthopaedic Surgery, New York University School of Medicine, NY, New York, United States.
| |
Collapse
|
38
|
Nagao M, Tanabe N, Manaka S, Naito M, Sekino J, Takayama T, Kawato T, Torigoe G, Kato S, Tsukune N, Maeno M, Suzuki N, Sato S. LIPUS suppressed LPS-induced IL-1α through the inhibition of NF-κB nuclear translocation via AT1-PLCβ pathway in MC3T3-E1 cells. J Cell Physiol 2017; 232:3337-3346. [PMID: 28063227 DOI: 10.1002/jcp.25777] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2016] [Accepted: 01/05/2017] [Indexed: 12/17/2022]
Abstract
Inflammatory cytokines, interleukin (IL)-1, IL-6, and TNF-α, are involved in inflammatory bone diseases such as rheumatoid osteoarthritis and periodontal disease. Particularly, periodontal disease, which destroys alveolar bone, is stimulated by lipopolysaccharide (LPS). Low-intensity pulsed ultrasound (LIPUS) is used for bone healing in orthopedics and dental treatments. However, the mechanism underlying effects of LIPUS on LPS-induced inflammatory cytokine are not well understood. We therefore aimed to investigate the role of LIPUS on LPS-induced IL-1α production. Mouse calvaria osteoblast-like cells MC3T3-E1 were incubated in the presence or absence of LPS (Porphyromonas gingivalis), and then stimulated with LIPUS for 30 min/day. To investigate the role of LIPUS, we determined the expression of IL-1α stimulated with LIPUS and treated with an angiotensin II receptor type 1 (AT1) antagonist, Losartan. We also investigate to clarify the pathway of LIPUS, we transfected siRNA silencing AT1 (siAT1) in MC3T3-E1. LIPUS inhibited mRNA and protein expression of LPS-induced IL-1α. LIPUS also reduced the nuclear translocation of NF-κB by LPS-induced IL-1α. Losartan and siAT1 blocked all the stimulatory effects of LIPUS on IL-1α production and IL-1α-mediated NF-κB translocation induced by LPS. Furthermore, PLCβ inhibitor U73122 recovered NF-κB translocation. These results suggest that LIPUS inhibits LPS-induced IL-1α via AT1-PLCβ in osteoblasts. We exhibit that these findings are in part of the signaling pathway of LIPUS on the anti-inflammatory effects of IL-1α expression.
Collapse
Affiliation(s)
- Mayu Nagao
- Nihon University Graduate School of Dentistry, Tokyo, Japan
| | - Natsuko Tanabe
- Department of Biochemistry, Nihon University Graduate School of Dentistry, Tokyo, Japan.,Division of Advanced Dental Treatment, Dental Research Center, Nihon University Graduate School of Dentistry, Tokyo, Japan
| | - Soichiro Manaka
- Department of Periodontology, Nihon University Graduate School of Dentistry, Tokyo, Japan
| | - Masako Naito
- Division of Advanced Dental Treatment, Dental Research Center, Nihon University Graduate School of Dentistry, Tokyo, Japan.,Department of Anatomy, Nihon University Graduate School of Dentistry, Tokyo, Japan
| | - Jumpei Sekino
- Nihon University Graduate School of Dentistry, Tokyo, Japan
| | - Tadahiro Takayama
- Department of Periodontology, Nihon University Graduate School of Dentistry, Tokyo, Japan.,Division of Functional Morphology, Dental Research Center, Nihon University Graduate School of Dentistry, Tokyo, Japan
| | - Takayuki Kawato
- Division of Advanced Dental Treatment, Dental Research Center, Nihon University Graduate School of Dentistry, Tokyo, Japan.,Department of Oral Health Sciences, Nihon University Graduate School of Dentistry, Tokyo, Japan
| | - Go Torigoe
- Nihon University Graduate School of Dentistry, Tokyo, Japan
| | | | - Naoya Tsukune
- Nihon University Graduate School of Dentistry, Tokyo, Japan
| | - Masao Maeno
- Division of Advanced Dental Treatment, Dental Research Center, Nihon University Graduate School of Dentistry, Tokyo, Japan.,Department of Oral Health Sciences, Nihon University Graduate School of Dentistry, Tokyo, Japan
| | - Naoto Suzuki
- Department of Biochemistry, Nihon University Graduate School of Dentistry, Tokyo, Japan.,Division of Advanced Dental Treatment, Dental Research Center, Nihon University Graduate School of Dentistry, Tokyo, Japan
| | - Shuichi Sato
- Department of Periodontology, Nihon University Graduate School of Dentistry, Tokyo, Japan.,Division of Functional Morphology, Dental Research Center, Nihon University Graduate School of Dentistry, Tokyo, Japan
| |
Collapse
|
39
|
HANMOTO T, TABUCHI Y, IKEGAME M, KONDO T, KITAMURA KI, ENDO M, KOBAYASHI I, MISHIMA H, SEKIGUCHI T, URATA M, SEKI A, YANO S, HATTORI A, SUZUKI N. Effects of low-intensity pulsed ultrasound on osteoclasts: Analysis with goldfish scales as a model of bone . Biomed Res 2017; 38:71-77. [DOI: 10.2220/biomedres.38.71] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Affiliation(s)
- Taizo HANMOTO
- Noto Marine Laboratory, Division of Marine Environmental Studies, Institute of Nature and Environmental Technology, Kanazawa University
| | - Yoshiaki TABUCHI
- Division of Molecular Genetics Research, Life Science Research Center, University of Toyama
| | - Mika IKEGAME
- Department of Oral Morphology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences
- ARCOCS, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences
| | - Takashi KONDO
- Department of Radiological Sciences, Gradu-ate School of Medicine and Pharmaceutical Sciences, University of Toyama
| | - Kei-ichiro KITAMURA
- Department of Clini-cal Laboratory Science, Division of Health Sciences, Graduate School of Medical Science, Kanazawa University
| | - Masato ENDO
- Department of Marine Biosciences, Tokyo University of Marine Science and Technology
| | - Isao KOBAYASHI
- Faculty of Natural System, Institute of Science and Engineering, Kanazawa University
| | | | - Toshio SEKIGUCHI
- Noto Marine Laboratory, Division of Marine Environmental Studies, Institute of Nature and Environmental Technology, Kanazawa University
| | - Makoto URATA
- Noto Marine Laboratory, Division of Marine Environmental Studies, Institute of Nature and Environmental Technology, Kanazawa University
- Institute of Noto SATOUMI Education and Studies
| | | | | | - Atsuhiko HATTORI
- Department of Biology, College of Liberal Arts and Sciences, Tokyo Medical and Dental University
| | - Nobuo SUZUKI
- Noto Marine Laboratory, Division of Marine Environmental Studies, Institute of Nature and Environmental Technology, Kanazawa University
| |
Collapse
|
40
|
Wang F, Li Y, Yang Z, Lu K, Zuo J, Zhou Z. Effect of Low-Intensity Pulsed Ultrasound on a Rat Model of Dentin-Dental Pulp Injury and Repair. ULTRASOUND IN MEDICINE & BIOLOGY 2017; 43:163-175. [PMID: 27814934 DOI: 10.1016/j.ultrasmedbio.2016.08.023] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/08/2016] [Revised: 08/17/2016] [Accepted: 08/18/2016] [Indexed: 06/06/2023]
Abstract
This study investigated histopathologic changes in dental pulp after treatment with low-intensity pulsed ultrasound (LIPUS). Fifty rats were randomly divided into an experimental group (n = 25) and a blank control group (n = 25). In the experimental group, a cavity was prepared in the bilateral maxillary first molars. The upper right first molars were stimulated with LIPUS (30 mW/cm2, 1.5 MHz) for 20 min/d. The cavities prepared in the left teeth were used as experimental controls (i.e., no LIPUS). Five rats in each group were sacrificed at days 1, 3, 5, 7 and 14. Inflammatory response was visible at different time points after cavity preparation, peaking at day 3, after which it gradually weakened. More reparative dentin was found on the LIPUS treatment side. transforming growth factor-β1 expression increased after treatment, peaking at day 5 and returning to normal at day 14 on both sides, but was stronger with LIPUS treatment. SMAD2 and SMAD3 expressions in the dental pulp gradually increased after cavity preparation, especially in the experimental group. LIPUS promoted the repair of dentin-pulp complex injury, to a certain extent and should be investigated further as a potential therapy.
Collapse
Affiliation(s)
- Fei Wang
- The College of Stomatology, Chongqing Medical University, Chongqing, China; Chongqing Key Laboratory for Oral Diseases and Biomedical Science, Chongqing, China; and Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, People's Republic of China
| | - Yueheng Li
- The College of Stomatology, Chongqing Medical University, Chongqing, China; Chongqing Key Laboratory for Oral Diseases and Biomedical Science, Chongqing, China; and Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, People's Republic of China
| | - Zhengyan Yang
- The College of Stomatology, Chongqing Medical University, Chongqing, China; Chongqing Key Laboratory for Oral Diseases and Biomedical Science, Chongqing, China; and Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, People's Republic of China
| | - Keke Lu
- The College of Stomatology, Chongqing Medical University, Chongqing, China; Chongqing Key Laboratory for Oral Diseases and Biomedical Science, Chongqing, China; and Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, People's Republic of China
| | - Jing Zuo
- The College of Stomatology, Chongqing Medical University, Chongqing, China; Chongqing Key Laboratory for Oral Diseases and Biomedical Science, Chongqing, China; and Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, People's Republic of China
| | - Zhi Zhou
- The College of Stomatology, Chongqing Medical University, Chongqing, China; Chongqing Key Laboratory for Oral Diseases and Biomedical Science, Chongqing, China; and Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, People's Republic of China.
| |
Collapse
|
41
|
Nagao M, Tanabe N, Manaka S, Takayama T, Kawato T, Torigoe G, Sekino J, Tsukune N, Ozaki M, Maeno M, Suzuki N, Sato S. Low-intensity pulsed ultrasound inhibits lipopolysaccharide-induced IL-6 and RANKL expression in osteoblasts. J Oral Sci 2017. [DOI: 10.2334/josnusd.16-0624] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022]
Affiliation(s)
- Mayu Nagao
- Division of Applied Oral Science, Nihon University Graduate School of Dentistry
| | - Natsuko Tanabe
- Department of Biochemistry, Nihon University School of Dentistry
- Division of Functional Morphology, Dental Research Center, Nihon University School of Dentistry
| | - Soichiro Manaka
- Department of Periodontology, Nihon University School of Dentistry
| | - Tadahiro Takayama
- Department of Periodontology, Nihon University School of Dentistry
- Division of Advanced Dental Treatment, Dental Research Center, Nihon University School of Dentistry
| | - Takayuki Kawato
- Division of Functional Morphology, Dental Research Center, Nihon University School of Dentistry
- Department of Oral Health Sciences, Nihon University School of Dentistry
| | - Go Torigoe
- Division of Applied Oral Science, Nihon University Graduate School of Dentistry
| | - Jumpei Sekino
- Division of Applied Oral Science, Nihon University Graduate School of Dentistry
| | - Naoya Tsukune
- Division of Applied Oral Science, Nihon University Graduate School of Dentistry
| | - Manami Ozaki
- Division of Applied Oral Science, Nihon University Graduate School of Dentistry
| | - Masao Maeno
- Division of Functional Morphology, Dental Research Center, Nihon University School of Dentistry
- Department of Oral Health Sciences, Nihon University School of Dentistry
| | - Naoto Suzuki
- Department of Biochemistry, Nihon University School of Dentistry
- Division of Functional Morphology, Dental Research Center, Nihon University School of Dentistry
| | - Shuichi Sato
- Department of Periodontology, Nihon University School of Dentistry
- Division of Advanced Dental Treatment, Dental Research Center, Nihon University School of Dentistry
| |
Collapse
|
42
|
Yamaguchi S, Aoyama T, Ito A, Nagai M, Iijima H, Tajino J, Zhang X, Wataru K, Kuroki H. Effect of Low-Intensity Pulsed Ultrasound after Mesenchymal Stromal Cell Injection to Treat Osteochondral Defects: An In Vivo Study. ULTRASOUND IN MEDICINE & BIOLOGY 2016; 42:2903-2913. [PMID: 27600474 DOI: 10.1016/j.ultrasmedbio.2016.07.021] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/05/2016] [Revised: 07/03/2016] [Accepted: 07/25/2016] [Indexed: 06/06/2023]
Abstract
We investigated the effect of low-intensity pulsed ultrasound (LIPUS) treatment combined with mesenchymal stromal cell (MSC) injection for cartilage repair and subchondral bone reconstitution for treatment of osteochondral defects. An osteochondral defect was created on both femur grooves of Wistar rats. Four weeks later, bone marrow MSCs were injected into the right knee joint. The rats were divided into two intervention groups: without or with LIPUS irradiation. Cartilage repair was evaluated histologically based on the Wakitani cartilage repair score. Subchondral bone reconstitution was evaluated as bone volume (BV)/tissue volume (TV) by micro-computed tomography analysis. MSC injection improved the cartilage repair score, and LIPUS irradiation improved BV/TV. Combination treatment promoted both cartilage repair and BV/TV improvement. Thus, MSC injection combined with LIPUS irradiation is more effective than either treatment alone in promoting concurrent cartilage repair and subchondral reconstitution.
Collapse
Affiliation(s)
- Shoki Yamaguchi
- Department of Motor Function Analysis, Human Health Sciences, Graduate School of Medicine, Kyoto University, Kyoto, Japan; Japan Society for the Promotion of Science, Tokyo, Japan
| | - Tomoki Aoyama
- Department of Development and Rehabilitation of Motor Function, Human Health Sciences, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Akira Ito
- Japan Society for the Promotion of Science, Tokyo, Japan; Department of Orthopaedic Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Momoko Nagai
- Congenital Anomaly Research Center, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Hirotaka Iijima
- Department of Motor Function Analysis, Human Health Sciences, Graduate School of Medicine, Kyoto University, Kyoto, Japan; Japan Society for the Promotion of Science, Tokyo, Japan
| | - Junichi Tajino
- Department of Motor Function Analysis, Human Health Sciences, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Xiangkai Zhang
- Department of Motor Function Analysis, Human Health Sciences, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Kiyan Wataru
- Department of Motor Function Analysis, Human Health Sciences, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Hiroshi Kuroki
- Department of Motor Function Analysis, Human Health Sciences, Graduate School of Medicine, Kyoto University, Kyoto, Japan.
| |
Collapse
|
43
|
Suzuki N, Hanmoto T, Yano S, Furusawa Y, Ikegame M, Tabuchi Y, Kondo T, Kitamura KI, Endo M, Yamamoto T, Sekiguchi T, Urata M, Mikuni-Takagaki Y, Hattori A. Low-intensity pulsed ultrasound induces apoptosis in osteoclasts: Fish scales are a suitable model for the analysis of bone metabolism by ultrasound. Comp Biochem Physiol A Mol Integr Physiol 2016; 195:26-31. [DOI: 10.1016/j.cbpa.2016.01.022] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Revised: 01/28/2016] [Accepted: 01/29/2016] [Indexed: 11/24/2022]
|
44
|
LIPUS promotes spinal fusion coupling proliferation of type H microvessels in bone. Sci Rep 2016; 6:20116. [PMID: 26830666 PMCID: PMC4735589 DOI: 10.1038/srep20116] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2015] [Accepted: 12/29/2015] [Indexed: 01/17/2023] Open
Abstract
Low-intensity pulsed ultrasound (LIPUS) has been found to accelerate spinal fusion. Type H microvessels are found in close relation with bone development. We analyzed the role of type H vessels in rat spinal fusion model intervened by LIPUS. It was found LIPUS could significantly accelerate bone fusion rate and enlarge bone callus. Osteoblasts were specifically located on the bone meshwork of the allograft, and were surrounded by type H microvessels. LIPUS could significantly increase the quantity of osteoblasts during spine fusion, which process was coupled with elevated angiogenesis of type H microvessels. Our results suggest that LIPUS may be a noninvasive adjuvant treatment modality in spinal fusion for clinical use. The treatment is recommended for usage for at least one month.
Collapse
|
45
|
Sommer AP. A mechanism for ultrasound/light-induced biostimulation. ANNALS OF TRANSLATIONAL MEDICINE 2015; 3:291. [PMID: 26697451 DOI: 10.3978/j.issn.2305-5839.2015.09.18] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Affiliation(s)
- Andrei P Sommer
- Institute of Micro and Nanomaterials, University of Ulm, 89081 Ulm, Germany
| |
Collapse
|