1
|
Mahadeo CO, Shahin-Shamsabadi A, Khodamoradi M, Fahnestock M, Selvaganapathy PR. The Effects of Electrical Stimulation on a 3D Osteoblast Cell Model. Cells 2025; 14:396. [PMID: 40136645 PMCID: PMC11941504 DOI: 10.3390/cells14060396] [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: 12/09/2024] [Revised: 01/31/2025] [Accepted: 03/04/2025] [Indexed: 03/27/2025] Open
Abstract
Electrical stimulation has been used with tissue engineering-based models to develop three-dimensional (3D), dynamic, research models that are more physiologically relevant than static two-dimensional (2D) cultures. For bone tissue, the effect of electrical stimulation has focused on promoting healing and regeneration of tissue to prevent bone loss. However, electrical stimulation can also potentially affect mature bone parenchymal cells such as osteoblasts to guide bone formation and the secretion of paracrine or endocrine factors. Due to a lack of physiologically relevant models, these phenomena have not been studied in detail. In vitro electrical stimulation models can be useful for gaining an understanding of bone physiology and its effects on paracrine tissues under different physiological and pathological conditions. Here, we use a 3D, dynamic, in vitro model of bone to study the effects of electrical stimulation conditions on protein and gene expression of SaOS-2 human osteosarcoma osteoblast-like cells. We show that different stimulation regimens, including different frequencies, exposure times, and stimulation patterns, can have different effects on the expression and secretion of the osteoblastic markers alkaline phosphatase and osteocalcin. These results reveal that electrical stimulation can potentially be used to guide osteoblast gene and protein expression.
Collapse
Affiliation(s)
- Crystal O. Mahadeo
- Neuroscience Graduate Program, McMaster University, Hamilton, ON L8S 4K1, Canada;
| | - Alireza Shahin-Shamsabadi
- School of Biomedical Engineering, McMaster University, Hamilton, ON L8S 4L8, Canada; (A.S.-S.); (M.K.)
- Department of Mechanical Engineering, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Maedeh Khodamoradi
- School of Biomedical Engineering, McMaster University, Hamilton, ON L8S 4L8, Canada; (A.S.-S.); (M.K.)
| | - Margaret Fahnestock
- Department of Psychiatry and Behavioural Neurosciences, McMaster University, Hamilton, ON L8S 4K1, Canada
| | - Ponnambalam Ravi Selvaganapathy
- School of Biomedical Engineering, McMaster University, Hamilton, ON L8S 4L8, Canada; (A.S.-S.); (M.K.)
- Department of Mechanical Engineering, McMaster University, Hamilton, ON L8S 4L8, Canada
| |
Collapse
|
2
|
Iwaki T, Sawaji Y, Masaoka T, Fukada E, Date M, Yamamoto K. Investigation of the effectiveness of intermittent electromagnetic field stimulation for early internal cartilaginous ossification in prechondrocytic ATDC5 cells. Bioelectromagnetics 2024; 45:226-234. [PMID: 38546158 DOI: 10.1002/bem.22501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 12/15/2023] [Accepted: 01/14/2024] [Indexed: 06/18/2024]
Abstract
Pulsed electromagnetic field (PEMF) stimulation has been widely applied clinically to promote bone healing; however, its detailed mechanism of action, particularly in endochondral ossification, remains elusive, and long-term stimulation is required for its satisfactory effect. The aim of this study was to investigate the involvement of the mammalian target of rapamycin (mTOR) pathway in chondrocyte differentiation and proliferation using a mouse prechondroblast cell line (ATDC5), and establish an efficient PEMF stimulation strategy for endochondral ossification. The changes in cell differentiation (gene expression levels of aggrecan, type II collagen, and type X collagen) and proliferation (cellular uptake of bromodeoxyuridine [BrdU]) in ATDC5 cells in the presence or absence of rapamycin, an mTOR inhibitor, was measured. The effects of continuous and intermittent PEMF stimulation on changes in cell differentiation and proliferation were compared. Rapamycin significantly suppressed the induction of cell differentiation markers and the cell proliferation activity. Furthermore, only intermittent PEMF stimulation continuously activated the mTOR pathway in ATDC5 cells, significantly promoting cell proliferation. These results demonstrate the involvement of the mTOR pathway in chondrocyte differentiation and proliferation and suggest that intermittent PEMF stimulation could be effective as a stimulus for endochondral ossification during fracture healing process, thereby reducing stimulation time.
Collapse
Affiliation(s)
- Takahiro Iwaki
- Department of Orthopedic Surgery, Tokyo Medical University, Tokyo, Japan
| | - Yasunobu Sawaji
- Department of Orthopedic Surgery, Tokyo Medical University, Tokyo, Japan
| | - Toshinori Masaoka
- Department of Orthopedic Surgery, Tokyo Medical University, Tokyo, Japan
| | - Eiichi Fukada
- Laboratory of piezoelectricity, Kobayasi Institute of Physical Research, Tokyo, Japan
| | - Munehiro Date
- Laboratory of piezoelectricity, Kobayasi Institute of Physical Research, Tokyo, Japan
| | - Kengo Yamamoto
- Department of Orthopedic Surgery, Tokyo Medical University, Tokyo, Japan
| |
Collapse
|
3
|
Franco-Obregón A. Harmonizing Magnetic Mitohormetic Regenerative Strategies: Developmental Implications of a Calcium-Mitochondrial Axis Invoked by Magnetic Field Exposure. Bioengineering (Basel) 2023; 10:1176. [PMID: 37892906 PMCID: PMC10604793 DOI: 10.3390/bioengineering10101176] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 10/05/2023] [Accepted: 10/08/2023] [Indexed: 10/29/2023] Open
Abstract
Mitohormesis is a process whereby mitochondrial stress responses, mediated by reactive oxygen species (ROS), act cumulatively to either instill survival adaptations (low ROS levels) or to produce cell damage (high ROS levels). The mitohormetic nature of extremely low-frequency electromagnetic field (ELF-EMF) exposure thus makes it susceptible to extraneous influences that also impinge on mitochondrial ROS production and contribute to the collective response. Consequently, magnetic stimulation paradigms are prone to experimental variability depending on diverse circumstances. The failure, or inability, to control for these factors has contributed to the existing discrepancies between published reports and in the interpretations made from the results generated therein. Confounding environmental factors include ambient magnetic fields, temperature, the mechanical environment, and the conventional use of aminoglycoside antibiotics. Biological factors include cell type and seeding density as well as the developmental, inflammatory, or senescence statuses of cells that depend on the prior handling of the experimental sample. Technological aspects include magnetic field directionality, uniformity, amplitude, and duration of exposure. All these factors will exhibit manifestations at the level of ROS production that will culminate as a unified cellular response in conjunction with magnetic exposure. Fortunately, many of these factors are under the control of the experimenter. This review will focus on delineating areas requiring technical and biological harmonization to assist in the designing of therapeutic strategies with more clearly defined and better predicted outcomes and to improve the mechanistic interpretation of the generated data, rather than on precise applications. This review will also explore the underlying mechanistic similarities between magnetic field exposure and other forms of biophysical stimuli, such as mechanical stimuli, that mutually induce elevations in intracellular calcium and ROS as a prerequisite for biological outcome. These forms of biophysical stimuli commonly invoke the activity of transient receptor potential cation channel classes, such as TRPC1.
Collapse
Affiliation(s)
- Alfredo Franco-Obregón
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore; ; Tel.: +65-6777-8427 or +65-6601-6143
- Institute of Health Technology and Innovation (iHealthtech), National University of Singapore, Singapore 117599, Singapore
- Biolonic Currents Electromagnetic Pulsing Systems Laboratory (BICEPS), National University of Singapore, Singapore 117599, Singapore
- NUS Centre for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
- Healthy Longevity Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117593, Singapore
- Nanomedicine Translational Research Programme, Centre for NanoMedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117544, Singapore
| |
Collapse
|
4
|
Zhang T, Zhao Z, Wang T. Pulsed electromagnetic fields as a promising therapy for glucocorticoid-induced osteoporosis. Front Bioeng Biotechnol 2023; 11:1103515. [PMID: 36937753 PMCID: PMC10020513 DOI: 10.3389/fbioe.2023.1103515] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Accepted: 02/21/2023] [Indexed: 03/06/2023] Open
Abstract
Glucocorticoid-induced osteoporosis (GIOP) is considered the third type of osteoporosis and is accompanied by high morbidity and mortality. Long-term usage of glucocorticoids (GCs) causes worsened bone quality and low bone mass via their effects on bone cells. Currently, there are various clinical pharmacological treatments to regulate bone mass and skeletal health. Pulsed electromagnetic fields (PEMFs) are applied to treat patients suffering from delayed fracture healing and non-unions. PEMFs may be considered a potential and side-effect-free therapy for GIOP. PEMFs inhibit osteoclastogenesis, stimulate osteoblastogenesis, and affect the activity of bone marrow mesenchymal stem cells (BMSCs), osteocytes and blood vessels, ultimately leading to the retention of bone mass and strength. However, the underlying signaling pathways via which PEMFs influence GIOP remain unclear. This review attempts to summarize the underlying cellular mechanisms of GIOP. Furthermore, recent advances showing that PEMFs affect bone cells are discussed. Finally, we discuss the possibility of using PEMFs as therapy for GIOP.
Collapse
Affiliation(s)
- Tianxiao Zhang
- Innovation Center for Wound Repair, West China Hospital, Sichuan University, Chengdu, China
| | - Zhiliang Zhao
- Innovation Center for Wound Repair, West China Hospital, Sichuan University, Chengdu, China
| | - Tiantian Wang
- Key Laboratory of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Institute of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- *Correspondence: Tiantian Wang,
| |
Collapse
|
5
|
Yan Z, Wang D, Cai J, Shen L, Jiang M, Liu X, Huang J, Zhang Y, Luo E, Jing D. High-specificity protection against radiation-induced bone loss by a pulsed electromagnetic field. SCIENCE ADVANCES 2022; 8:eabq0222. [PMID: 36001662 PMCID: PMC9401628 DOI: 10.1126/sciadv.abq0222] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 07/11/2022] [Indexed: 05/28/2023]
Abstract
Radiotherapy increases tumor cure and survival rates; however, radiotherapy-induced bone damage remains a common issue for which effective countermeasures are lacking, especially considering tumor recurrence risks. We report a high-specificity protection technique based on noninvasive electromagnetic field (EMF). A unique pulsed-burst EMF (PEMF) at 15 Hz and 2 mT induces notable Ca2+ oscillations with robust Ca2+ spikes in osteoblasts in contrast to other waveforms. This waveform parameter substantially inhibits radiotherapy-induced bone loss by specifically modulating osteoblasts without affecting other bone cell types or tumor cells. Mechanistically, primary cilia are identified as major PEMF sensors in osteoblasts, and the differentiated ciliary expression dominates distinct PEMF sensitivity between osteoblasts and tumor cells. PEMF-induced unique Ca2+ oscillations depend on interactions between ciliary polycystins-1/2 and endoplasmic reticulum, which activates the Ras/MAPK/AP-1 axis and subsequent DNA repair Ku70 transcription. Our study introduces a previously unidentified method against radiation-induced bone damage in a noninvasive, cost-effective, and highly specific manner.
Collapse
Affiliation(s)
- Zedong Yan
- Department of Biomedical Engineering, Fourth Military Medical University, Xi’an, China
| | - Dan Wang
- Department of Biomedical Engineering, Fourth Military Medical University, Xi’an, China
| | - Jing Cai
- College of Basic Medicine, Shaanxi University of Chinese Medicine, Xianyang, China
| | - Liangliang Shen
- The State Key Laboratory of Cancer Biology, Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi’an, China
| | - Maogang Jiang
- Department of Biomedical Engineering, Fourth Military Medical University, Xi’an, China
| | - Xiyu Liu
- Department of Biomedical Engineering, Fourth Military Medical University, Xi’an, China
| | - Jinghui Huang
- Institute of Orthopaedics, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Yong Zhang
- Department of Pulmonary and Critical Care of Medicine, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Erping Luo
- Department of Biomedical Engineering, Fourth Military Medical University, Xi’an, China
| | - Da Jing
- Department of Biomedical Engineering, Fourth Military Medical University, Xi’an, China
- The Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, Fourth Military Medical University, Xi’an, China
- Shaanxi Provincial Key Laboratory of Bioelectromagnetic Detection and Intelligent Perception, Fourth Military Medical University, Xi’an, China
| |
Collapse
|
6
|
Effects of Peripheral Electromagnetic Fields on Spasticity: A Systematic Review. J Clin Med 2022; 11:jcm11133739. [PMID: 35807019 PMCID: PMC9267146 DOI: 10.3390/jcm11133739] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 06/22/2022] [Accepted: 06/23/2022] [Indexed: 02/05/2023] Open
Abstract
Electromagnetic fields are emerging as a therapeutic option for patients with spasticity. They have been applied at brain or peripheral level. The effects of electromagnetic fields applied to the brain have been extensively studied for years in spasticity, but not so at the peripheral level. Therefore, the purpose of our work is to analyze the effects of electromagnetic fields, applied peripherally to spasticity. A systematic review was conducted resulting in 10 clinical trials. The frequency ranged from 1 Hz to 150 Hz, with 25 Hz being the most commonly used and the intensity it was gradually increased but there was low homogeneity in how it was increased. Positive results on spasticity were found in 80% of the studies: improvements in stretch reflex threshold, self questionnaire about difficulties related to spasticity, clinical spasticity score, performance scale, Ashworth scale, spastic tone, Hmax/Mmax Ratio and active and passive dorsal flexion. However, results must be taken with caution due to the large heterogeneity and the small number of articles. In future studies, it would be interesting to agree on the parameters to be used, as well as the way of assessing spasticity, to be more objective in the study of their effectiveness.
Collapse
|
7
|
Montorsi M, Genchi GG, De Pasquale D, De Simoni G, Sinibaldi E, Ciofani G. Design, Fabrication, and Characterization of a Multimodal Reconfigurable Bioreactor for Bone Tissue Engineering. Biotechnol Bioeng 2022; 119:1965-1979. [PMID: 35383894 PMCID: PMC9324218 DOI: 10.1002/bit.28100] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 03/15/2022] [Accepted: 03/31/2022] [Indexed: 11/18/2022]
Abstract
In the past decades, bone tissue engineering developed and exploited many typologies of bioreactors, which, besides providing proper culture conditions, aimed at integrating those bio‐physical stimulations that cells experience in vivo, to promote osteogenic differentiation. Nevertheless, the highly challenging combination and deployment of many stimulation systems into a single bioreactor led to the generation of several unimodal bioreactors, investigating one or at mostly two of the required biophysical stimuli. These systems miss the physiological mimicry of bone cells environment, and often produced contrasting results, thus making the knowledge of bone mechanotransduction fragmented and often inconsistent. To overcome this issue, in this study we developed a perfusion and electroactive‐vibrational reconfigurable stimulation bioreactor to investigate the differentiation of SaOS‐2 bone‐derived cells, hosting a piezoelectric nanocomposite membrane as cell culture substrate. This multimodal perfusion bioreactor is designed based on a numerical (finite element) model aimed at assessing the possibility to induce membrane nano‐scaled vibrations (with ~12 nm amplitude at a frequency of 939 kHz) during perfusion (featuring 1.46 dyn cm−2 wall shear stress), large enough for inducing a physiologically‐relevant electric output (in the order of 10 mV on average) on the membrane surface. This study explored the effects of different stimuli individually, enabling to switch on one stimulation at a time, and then to combine them to induce a faster bone matrix deposition rate. Biological results demonstrate that the multimodal configuration is the most effective in inducing SaOS‐2 cell differentiation, leading to 20‐fold higher collagen deposition compared to static cultures, and to 1.6‐ and 1.2‐fold higher deposition than the perfused‐ or vibrated‐only cultures. These promising results can provide tissue engineering scientists with a comprehensive and biomimetic stimulation platform for a better understanding of mechanotransduction phenomena beyond cells differentiation.
Collapse
Affiliation(s)
- Margherita Montorsi
- Istituto Italiano di Tecnologia, Smart Bio-Interfaces, Viale Rinaldo Piaggio 34, 56025, Pontedera, Italy.,Scuola Superiore Sant'Anna, The BioRobotics Institute, Viale Rinaldo Piaggio 34, 56025, Pontedera, Italy
| | - Giada Graziana Genchi
- Istituto Italiano di Tecnologia, Smart Bio-Interfaces, Viale Rinaldo Piaggio 34, 56025, Pontedera, Italy
| | - Daniele De Pasquale
- Istituto Italiano di Tecnologia, Smart Bio-Interfaces, Viale Rinaldo Piaggio 34, 56025, Pontedera, Italy
| | - Giorgio De Simoni
- CNR, Nanoscience Institute, NEST Laboratory, Piazza San Silvestro 12, 56127, Pisa, Italy
| | - Edoardo Sinibaldi
- Istituto Italiano di Tecnologia, Bioinspired Soft Robotics, Via Morego 30, 16163, Genova, Italy
| | - Gianni Ciofani
- Istituto Italiano di Tecnologia, Smart Bio-Interfaces, Viale Rinaldo Piaggio 34, 56025, Pontedera, Italy
| |
Collapse
|
8
|
Osteogenesis Modulation: Induction of Mandibular Bone Growth in Adults by Electrical Field for Aesthetic Purposes. Aesthetic Plast Surg 2022; 46:197-206. [PMID: 34622330 PMCID: PMC8831273 DOI: 10.1007/s00266-021-02600-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Accepted: 09/12/2021] [Indexed: 10/25/2022]
Abstract
BACKGROUND A new technique in plastic surgery termed Osteogenesis Modulation is described. This technique uses a surgically implanted, battery-operated medical device to deliver customized electrical pulses to produce mandibular bone growth. This device was designed to be a temporary, nonpermanent implant. The purpose of this study was to review both the safety and efficacy of Osteogenesis Modulation. METHODS This study comprises two phases. Phase I involved experimental technology development and animal experiments. Phase II included technology development for clinical use and a clinical trial. In Phase II, four patients with a diagnosis of mandibular hypoplasia and microgenia underwent surgical implantation of the novel medical device over the chin bone. Once a satisfactory change of contour of mandibular bone was achieved, the devices were removed. In all patients, the devices were left in place for 12 months, then surgically removed under local anesthesia. Preoperative and long-term postoperative cephalometric controls were done. RESULTS In all patients, symmetrical mandibular bone growth was observed with good-to-excellent aesthetic results. The overall follow-up period was 39 months. Cephalometric controls taken 3 to 6 months after the device removal showed an average increase in mandible length of 5.26mm (range, 2.83-7.60mm) CONCLUSIONS: Preliminary clinical results suggest that Osteogenesis Modulation is a safe, minimally invasive, and effective alternative treatment for the correction of mandibular hypoplasia in selected cases. LEVEL OF EVIDENCE IV This journal requires that authors assign a level of evidence to each article. For a full description of these Evidence-Based Medicine ratings, please refer to the Table of Contents or the online Instructions to Authors www.springer.com/00266 .
Collapse
|
9
|
Dastjerdi S, Malikan M, Akgöz B, Civalek Ö, Wiczenbach T, Eremeyev VA. On the deformation and frequency analyses of SARS-CoV-2 at nanoscale. INTERNATIONAL JOURNAL OF ENGINEERING SCIENCE 2022; 170:103604. [PMID: 34728858 PMCID: PMC8554078 DOI: 10.1016/j.ijengsci.2021.103604] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Accepted: 10/23/2021] [Indexed: 05/07/2023]
Abstract
The SARS-CoV-2 virus, which has emerged as a Covid-19 pandemic, has had the most significant impact on people's health, economy, and lifestyle around the world today. In the present study, the SARS-CoV-2 virus is mechanically simulated to obtain its deformation and natural frequencies. The virus under analysis is modeled on a viscoelastic spherical structure. The theory of shell structures in mechanics is used to derive the governing equations. Whereas the virus has nanometric size, using classical theories may give incorrect results. Consequently, the nonlocal elasticity theory is used to consider the effect of interatomic forces on the results. From the mechanical point of view, if a structure vibrates with a natural frequency specific to it, the resonance phenomenon will occur in that structure, leading to its destruction. Therefore, it is possible that the protein chains of SARS-CoV-2 would be destroyed by vibrating it at natural frequencies. Since the mechanical properties of SARS-CoV-2 are not clearly known due to the new emergence of this virus, deformation and natural frequencies are obtained in a specific interval. Researchers could also use this investigation as a pioneering study to find a non-vaccine treatment solution for the SARS-CoV-2 virus and various viruses, including HIV.
Collapse
Affiliation(s)
- Shahriar Dastjerdi
- Division of Mechanics, Civil Engineering Department, Akdeniz University, Antalya, Turkey
| | - Mohammad Malikan
- Department of Mechanics of Materials and Structures, Faculty of Civil and Environmental Engineering, Gdansk University of Technology, Gdansk, Poland
| | - Bekir Akgöz
- Division of Mechanics, Civil Engineering Department, Akdeniz University, Antalya, Turkey
| | - Ömer Civalek
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan
| | - Tomasz Wiczenbach
- Department of Mechanics of Materials and Structures, Faculty of Civil and Environmental Engineering, Gdansk University of Technology, Gdansk, Poland
| | - Victor A Eremeyev
- Department of Mechanics of Materials and Structures, Faculty of Civil and Environmental Engineering, Gdansk University of Technology, Gdansk, Poland
- Research and Education Center "Materials" Don State Technical University, Gagarina sq., 1, 344000 Rostov on Don, Russia
- DICAAR, Università degli Studi di Cagliari, Via Marengo, 2, 09123, Cagliari, Italy
| |
Collapse
|
10
|
Kim Y, Lim H, Lee E, Ki G, Seo Y. Synergistic effect of electromagnetic fields and nanomagnetic particles on osteogenesis through calcium channels and p-ERK signaling. J Orthop Res 2021; 39:1633-1646. [PMID: 33150984 PMCID: PMC8451839 DOI: 10.1002/jor.24905] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2020] [Revised: 10/21/2020] [Accepted: 10/31/2020] [Indexed: 02/04/2023]
Abstract
Electromagnetic fields (EMFs) are widely used in a number of cell therapies and bone disorder treatments, and nanomagnetic particles (NMPs) also promote cell activity. In this study, we investigated the synergistic effects of EMFs and NMPs on the osteogenesis of the human Saos-2 osteoblast cell line and in a rat calvarial defect model. The Saos-2 cells and critical-size calvarial defects of the rats were exposed to EMF (1 mT, 45 Hz, 8 h/day) with or without Fe3 O4 NMPs. Biocompatibility was evaluated with MTT (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide) and LDH (lactate dehydrogenase) assays. This analysis showed that NMP and EMF did not induce cell toxicity. Quantitative reverse-transcription polymerase chain reaction indicated that the osteogenesis-related markers were highly expressed in the NMP-incorporated Saos-2 cells after exposure to EMF. Also, the expression of gene-encoding proteins involved in calcium channels was activated and the calcium concentration of the NMP-incorporated + EMF-exposed group was increased compared with the control group. In particular, in the NMP-incorporated + EMF-exposed group, all osteogenic proteins were more abundantly expressed than in the control group. This indicated that the NMP incorporation + EMF exposure induced a signaling pathway through activation of p-ERK and calcium channels. Also, in vivo evaluation revealed that rat calvarial defects treated with EMFs and NMPs had good regeneration results with new bone formation and increased mineral density after 6 weeks. Altogether, these results suggest that NMP treatment or EMF exposure of Saos-2 cells can increase osteogenic activity and NMP incorporation following EMF exposure which is synergistically efficient for osteogenesis.
Collapse
Affiliation(s)
- Yu‐Mi Kim
- Department of Medical Biotechnology (BK21 Plus Team)Dongguk UniversityGoyang‐siKorea
| | - Han‐Moi Lim
- Department of Medical Biotechnology (BK21 Plus Team)Dongguk UniversityGoyang‐siKorea
| | - Eun‐Chul Lee
- Department of Medical Biotechnology (BK21 Plus Team)Dongguk UniversityGoyang‐siKorea
| | - Ga‐Eun Ki
- Department of Medical Biotechnology (BK21 Plus Team)Dongguk UniversityGoyang‐siKorea
| | - Young‐Kwon Seo
- Department of Medical Biotechnology (BK21 Plus Team)Dongguk UniversityGoyang‐siKorea
| |
Collapse
|
11
|
Mansourian M, Shanei A. Evaluation of Pulsed Electromagnetic Field Effects: A Systematic Review and Meta-Analysis on Highlights of Two Decades of Research In Vitro Studies. BIOMED RESEARCH INTERNATIONAL 2021; 2021:6647497. [PMID: 34368353 PMCID: PMC8342182 DOI: 10.1155/2021/6647497] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/04/2020] [Revised: 05/30/2021] [Accepted: 06/27/2021] [Indexed: 12/13/2022]
Abstract
Pulsed electromagnetic field (PEMF) therapy is a type of physical stimulation that affects biological systems by producing interfering or coherent fields. Given that cell types are significantly distinct, which represents an important factor in stimulation, and that PEMFs can have different effects in terms of frequency and intensity, time of exposure, and waveform. This study is aimed at investigating if distinct positive and negative responses would correspond to specific characteristics of cells, frequency and flux density, time of exposure, and waveform. Necessary data were abstracted from the experimental observations of cell-based in vitro models. The observations were obtained from 92 publications between the years 1999 and 2019, which are available on PubMed and Web of Science databases. From each of the included studies, type of cells, pulse frequency of exposure, exposure flux density, and assayed cell responses were extracted. According to the obtained data, most of the experiments were carried out on human cells, and out of 2421 human cell experiments, cell changes were observed only in 51.05% of the data. In addition, the results pointed out the potential effects of PEMFs on some human cell types such as MG-63 human osteosarcoma cells (p value < 0.001) and bone marrow mesenchymal stem cells. However, human osteogenic sarcoma SaOS-2 (p < 0.001) and human adipose-derived mesenchymal stem cells (AD-MSCs) showed less sensitivity to PEMFs. Nevertheless, the evidence suggests that frequencies higher than 100 Hz, flux densities between 1 and 10 mT, and chronic exposure more than 10 days would be more effective in establishing a cellular response. This study successfully reported useful information about the role of cell type and signal characteristic parameters, which were of high importance for targeted therapies using PEMFs. Our findings would provide a deeper understanding about the effect of PEMFs in vitro, which could be useful as a reference for many in vivo experiments or preclinical trials.
Collapse
Affiliation(s)
- Mahsa Mansourian
- Department of Medical Physics, Faculty of Medicine, Isfahan University of Medical Science, Isfahan, Hezar Jerib Avenue, Isfahan, Iran
| | - Ahmad Shanei
- Department of Medical Physics, Faculty of Medical Science, Isfahan University of Medical Science, Isfahan, Hezar Jerib Street, Isfahan, Iran
| |
Collapse
|
12
|
Parmaksiz M, Lalegül-Ülker Ö, Vurat MT, Elçin AE, Elçin YM. Magneto-sensitive decellularized bone matrix with or without low frequency-pulsed electromagnetic field exposure for the healing of a critical-size bone defect. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 124:112065. [PMID: 33947558 DOI: 10.1016/j.msec.2021.112065] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 02/14/2021] [Accepted: 03/12/2021] [Indexed: 12/12/2022]
Abstract
Bioactive ECM-based materials mimic the complex composition and structure of natural tissues. Decellularized cancellous bone matrix (DBM) has potential for guiding new bone formation and accelerating the regeneration process. On the other hand, low frequency-pulsed electromagnetic field (LF-PEMF) has been shown to enhance the regeneration capacity of bone defects. The present study sought to explore the feasibility of using DBM and DBM/MNP, and LF-PEMF for treating critical-size bone defects. Firstly, decellularization protocol was optimized to obtain a bioactive DBM, then MNPs were incorporated. Later, the physical, chemical and biological properties of DBM and DBM/MNP were assessed in vitro. MNPs homogeneously distributed into the DBM were not found to be toxic to human osteoblast cultures. Finally, an in vivo study was carried out with DBM and DBM/MNP composites in a bilateral critical-size rat cranial defect model (n = 48) with or without LF-PEMF exposure for 45 and 90 days. The histomorphometric and radiographic evaluations revealed that, while the collagen (positive control) and Sham (negative control) groups showed high incidence of fibrous connective tissue together with low level of osteogenic activity, both the DBM and DBM/MNP-grafted groups significantly promoted new bone tissue formation and angiogenesis, by the appropriate use of LF-PEMF for 90 days.
Collapse
Affiliation(s)
- Mahmut Parmaksiz
- Tissue Engineering, Biomaterials and Nanobiotechnology Laboratory, Ankara University Faculty of Science, Ankara University Stem Cell Institute, Ankara, Turkey
| | - Özge Lalegül-Ülker
- Tissue Engineering, Biomaterials and Nanobiotechnology Laboratory, Ankara University Faculty of Science, Ankara University Stem Cell Institute, Ankara, Turkey
| | - Murat Taner Vurat
- Tissue Engineering, Biomaterials and Nanobiotechnology Laboratory, Ankara University Faculty of Science, Ankara University Stem Cell Institute, Ankara, Turkey
| | - Ayşe Eser Elçin
- Tissue Engineering, Biomaterials and Nanobiotechnology Laboratory, Ankara University Faculty of Science, Ankara University Stem Cell Institute, Ankara, Turkey
| | - Yaşar Murat Elçin
- Tissue Engineering, Biomaterials and Nanobiotechnology Laboratory, Ankara University Faculty of Science, Ankara University Stem Cell Institute, Ankara, Turkey; Biovalda Health Technologies, Inc., Ankara, Turkey.
| |
Collapse
|
13
|
Tabatabai TS, Haji Ghasem Kashani M, Maskani R, Nasiri M, Nabavi Amri SA, Atashi A, Bitaraf FS. Synergic effects of extremely low-frequency electromagnetic field and betaine on in vitro osteogenic differentiation of human adipose tissue-derived mesenchymal stem cells. In Vitro Cell Dev Biol Anim 2021; 57:468-476. [PMID: 33770338 DOI: 10.1007/s11626-021-00558-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Accepted: 02/25/2021] [Indexed: 11/30/2022]
Abstract
Human adipose tissue-derived mesenchymal stem cells (hADSCs) due to easy extraction, relative abundance, in vitro expansion and differentiation potential, frozen storage capability, and ability to secrete cytokines, compared to other stem cells, are appropriate candidate in regenerative medicine. Extremely low-frequency electromagnetic fields (ELF-EMF) and betaine are two safe factors in bone lesions repair. This study was designed to assess the osteogenic differentiation potential of these factors on hADSCs. The samples were collected from women undergoing liposuction after obtaining written consent. The hADSCs were extracted and treated with osteogenesis differentiation medium (OD) as the positive control, with OD and betaine (BET group), with OD and EMF (EMF group), and with OD and betaine and EMF (BET+EMF group) for 21 d; the negative control consisted of cells without treatment. Betaine 10 mM and EMF with 50-Hz frequency, 1-mT intensity (8 h daily), and in the form of sinus wave were used. Osteogenic differentiation was evaluated by Alizarin Red staining, alkaline phosphatase activity, calcium deposition, and real-time PCR. A significant increase in calcium deposition in the BET+EMF group was observed compared to the other groups. The activity of alkaline phosphatase in the positive control and BET groups was increased significantly compared to EMF and BET + EMF groups and a significant increase of this enzyme activity in the BET + EMF compared to EMF group was observed. The expression of RUNX2 and OCN genes in the EMF-treated groups were significantly reduced compared to the non-EMF-treated groups, and BET+EMF showed a significant increase of RUNX2 gene expression as compared the EMF group. The ELF-EMF leads to a decrease in the osteogenic differentiation and the expression RUNX2 and OCN genes in hADSCs. But osteogenic differentiation and RUNX2 gene expression were increased post-induction by betaine. The synergic effect of betaine and EMF on the osteogenic differentiation and related genes expression of hADSCs was higher than EMF.
Collapse
Affiliation(s)
- Tayebeh Sadat Tabatabai
- Department of Cellular and Molecular Biology, School of Biology and Institute of Biological Sciences, Damghan University, Damghan, Iran
| | - Maryam Haji Ghasem Kashani
- Department of Cellular and Molecular Biology, School of Biology and Institute of Biological Sciences, Damghan University, Damghan, Iran.
| | - Reza Maskani
- Department of Medical Sciences, School of Paramedical, Shahroud University of Medical Sciences, Shahroud, Iran
| | - Meysam Nasiri
- Department of Cellular and Molecular Biology, School of Biology and Institute of Biological Sciences, Damghan University, Damghan, Iran
| | | | - Amir Atashi
- Department of Medical Sciences, School of Paramedical, Shahroud University of Medical Sciences, Shahroud, Iran
| | - Fateme Sadat Bitaraf
- Department of Medical Biotechnology, School of Medicine, Shahroud University of Medical Sciences, Shahroud, Iran
| |
Collapse
|
14
|
Gelmi A, Schutt CE. Stimuli-Responsive Biomaterials: Scaffolds for Stem Cell Control. Adv Healthc Mater 2021; 10:e2001125. [PMID: 32996270 PMCID: PMC11468740 DOI: 10.1002/adhm.202001125] [Citation(s) in RCA: 84] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 08/18/2020] [Indexed: 12/28/2022]
Abstract
Stem cell fate is closely intertwined with microenvironmental and endogenous cues within the body. Recapitulating this dynamic environment ex vivo can be achieved through engineered biomaterials which can respond to exogenous stimulation (including light, electrical stimulation, ultrasound, and magnetic fields) to deliver temporal and spatial cues to stem cells. These stimuli-responsive biomaterials can be integrated into scaffolds to investigate stem cell response in vitro and in vivo, and offer many pathways of cellular manipulation: biochemical cues, scaffold property changes, drug release, mechanical stress, and electrical signaling. The aim of this review is to assess and discuss the current state of exogenous stimuli-responsive biomaterials, and their application in multipotent stem cell control. Future perspectives in utilizing these biomaterials for personalized tissue engineering and directing organoid models are also discussed.
Collapse
Affiliation(s)
- Amy Gelmi
- School of ScienceCollege of Science, Engineering and HealthRMIT UniversityMelbourneVIC3001Australia
| | - Carolyn E. Schutt
- Department of Biomedical EngineeringKnight Cancer Institute Cancer Early Detection Advanced Research Center (CEDAR)Oregon Health and Science UniversityPortlandOR97201USA
| |
Collapse
|
15
|
Evaluation of pulsed electromagnetic field protocols in implant osseointegration: in vivo and in vitro study. Clin Oral Investig 2020; 25:2925-2937. [PMID: 33033921 DOI: 10.1007/s00784-020-03612-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Accepted: 09/29/2020] [Indexed: 10/23/2022]
Abstract
OBJECTIVES The present study aims to evaluate two protocols of pulsed electromagnetic field (PEMF) on osseointegration and establish one that addresses ideal parameters for its use in dentistry, especially in the optimization of the implants osseointegration process. MATERIALS AND METHODS Sixty male rats (Wistar) were allocated into three experimental groups: control (GC), test A (GTA, 3 h exposed), and test B (GTB, 1 h exposed). All animals received titanium implants in both tibias, and PEMF application (15 Hz, ± 1 mT, 5 days/week) occurred only in the test groups. They were euthanized at 03, 07, 21, and 45 days after PEMF therapy. Removal torque, histomorphometric measurements, three-dimensional radiographic evaluation, and in vitro biological assay analyses were performed. RESULTS GTB showed better results compared with GTA in removal torque tests, in bone volume and bone mineral density, cell viability, total protein content, and mineralization nodules (p < 0.05). GTA showed better performance in trabecular bone thickness and cell proliferation compared with GTB (p < 0.05), especially at osseointegration early periods. In the histomorphometric analysis and number of trabeculae, there were no differences in the test groups. CONCLUSION PEMF as a biostimulator was effective in optimizing the events in bone tissue that lead to osseointegration, especially when applied for a shorter time and in the initial periods of bone healing. CLINICAL RELEVANCE The PEMF therapy is an effective alternative method for optimizing bone healing.
Collapse
|
16
|
Cosoli G, Scalise L, De Leo A, Russo P, Tricarico G, Tomasini EP, Cerri G. Development of a Novel Medical Device for Mucositis and Peri-Implantitis Treatment. Bioengineering (Basel) 2020; 7:E87. [PMID: 32764313 PMCID: PMC7552606 DOI: 10.3390/bioengineering7030087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 07/14/2020] [Accepted: 08/03/2020] [Indexed: 11/24/2022] Open
Abstract
In spite of all the developments in dental implantology techniques, peri-implant diseases are frequent (prevalence up to 80% and 56% of subjects for mucositis and peri-implantitis, respectively) and there is an urgency for an effective treatment strategy. This paper presents an innovative electromedical device for the electromagnetic treatment of mucositis and peri-implantitis diseases. This device is also equipped with a measurement part for bioimpedance, which reflects the health conditions of a tissue, thus allowing clinicians to objectively detect impaired areas and to monitor the severity of the disease, evaluate the treatment efficacy, and adjust it accordingly. The design of the device was realized considering literature data, clinical evidence, numerical simulation results, and electromagnetic compatibility (EMC) pre-compliance tests, involving both clinicians and engineers, to better understand all the needs and translate them into design requirements. The reported system is being tested in more than 50 dental offices since 2019, providing efficient treatments for mucositis and peri-implantitis, with success rates of approximately 98% and 80%, respectively.
Collapse
Affiliation(s)
- Gloria Cosoli
- Department of Industrial Engineering and Mathematical Sciences, Università Politecnica delle Marche, 60131 Ancona, Italy; (L.S.); (E.P.T.)
| | - Lorenzo Scalise
- Department of Industrial Engineering and Mathematical Sciences, Università Politecnica delle Marche, 60131 Ancona, Italy; (L.S.); (E.P.T.)
| | - Alfredo De Leo
- Department of Information Engineering, Università Politecnica delle Marche, 60131 Ancona, Italy; (A.D.L.); (P.R.); (G.C.)
| | - Paola Russo
- Department of Information Engineering, Università Politecnica delle Marche, 60131 Ancona, Italy; (A.D.L.); (P.R.); (G.C.)
| | - Gerardo Tricarico
- Department of Health Sciences, Università del Piemonte Orientale, 28100 Novara, Italy;
| | - Enrico Primo Tomasini
- Department of Industrial Engineering and Mathematical Sciences, Università Politecnica delle Marche, 60131 Ancona, Italy; (L.S.); (E.P.T.)
| | - Graziano Cerri
- Department of Information Engineering, Università Politecnica delle Marche, 60131 Ancona, Italy; (A.D.L.); (P.R.); (G.C.)
| |
Collapse
|
17
|
In Vitro Production of Calcified Bone Matrix onto Wool Keratin Scaffolds via Osteogenic Factors and Electromagnetic Stimulus. MATERIALS 2020; 13:ma13143052. [PMID: 32650489 PMCID: PMC7411850 DOI: 10.3390/ma13143052] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 07/03/2020] [Accepted: 07/04/2020] [Indexed: 12/15/2022]
Abstract
Pulsed electromagnetic field (PEMF) has drawn attention as a potential tool to improve the ability of bone biomaterials to integrate into the surrounding tissue. We investigated the effects of PEMF (frequency, 75 Hz; magnetic induction amplitude, 2 mT; pulse duration, 1.3 ms) on human osteoblast-like cells (SAOS-2) seeded onto wool keratin scaffolds in terms of proliferation, differentiation, and production of the calcified bone extracellular matrix. The wool keratin scaffold offered a 3D porous architecture for cell guesting and nutrient diffusion, suggesting its possible use as a filler to repair bone defects. Here, the combined approach of applying a daily PEMF exposure with additional osteogenic factors stimulated the cells to increase both the deposition of bone-related proteins and calcified matrix onto the wool keratin scaffolds. Also, the presence of SAOS-2 cells, or PEMF, or osteogenic factors did not influence the compression behavior or the resilience of keratin scaffolds in wet conditions. Besides, ageing tests revealed that wool keratin scaffolds were very stable and showed a lower degradation rate compared to commercial collagen sponges. It is for these reasons that this tissue engineering strategy, which improves the osteointegration properties of the wool keratin scaffold, may have a promising application for long term support of bone formation in vivo.
Collapse
|
18
|
Naghibzadeh M, Gholampour S, Naghibzadeh M, Sadeghian-Nodoushan F, Nikukar H. The effect of electromagnetic field on decreasing and increasing of the growth and proliferation rate of dermal fibroblast cell. Dermatol Ther 2020; 33:e13803. [PMID: 32526050 DOI: 10.1111/dth.13803] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2020] [Revised: 04/06/2020] [Accepted: 06/06/2020] [Indexed: 11/29/2022]
Abstract
Maintaining the health of dermal fibroblast cells and controlling their growth and proliferation would directly affect the health of skin tissues. The present study encompassed three control and three experimental specimens, which were different in terms of the duration of exposure to electromagnetic field (EMF) and intensity. With a decrease in intensity from 2 to 1 mT during 24, 48, and 72 h after exposing the cells to EMF, the frequency of the sample fibroblast cells increased by 60.3%, 144.9%, and 90.1%, respectively. With an increase in intensity from 3 to 4 mT during 48 and 72 h of exposure to EMF, the frequencies of the sample fibroblast cells decreased by 6.8% and 86.7%, respectively. It seems to be possible to achieve the most desirable condition to help the restoration of wounds and skin lesions through decreasing the exposure intensity from 2 to 0.5 mT and increasing EMF exposure time from 24 to 72 h simultaneously and non-invasively. The most desirable approach to improve the treatment of skin cancers non-invasively is to increase the intensity from 3 to 5 mT and to enhance EMF exposure time from 48 to 72 h.
Collapse
Affiliation(s)
- Mehran Naghibzadeh
- Department of Biomedical Engineering, North Tehran Branch, Islamic Azad University, Tehran, Iran
| | - Seifollah Gholampour
- Department of Biomedical Engineering, North Tehran Branch, Islamic Azad University, Tehran, Iran
| | - Majid Naghibzadeh
- Medical Nanotechnology and Tissue Engineering Research Center, Yazd Reproductive Sciences Institute, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Fatemeh Sadeghian-Nodoushan
- Medical Nanotechnology and Tissue Engineering Research Center, Yazd Reproductive Sciences Institute, Shahid Sadoughi University of Medical Sciences, Yazd, Iran.,Department of Tissue engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Habib Nikukar
- Medical Nanotechnology and Tissue Engineering Research Center, Yazd Reproductive Sciences Institute, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| |
Collapse
|
19
|
Topal O, Çina Aksoy M, Ciriş İM, Doğuç DK, Sert S, Çömlekçi S. Assessment of the effect of pulsed electromagnetic field application on the healing of bone defects in rats with heparin-induced osteoporosis. Electromagn Biol Med 2020; 39:206-217. [PMID: 32419512 DOI: 10.1080/15368378.2020.1762636] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Osteoporosis is a systemic skeletal disease characterized by an increase in bone fragility and fracture risk due to low bone mass and deterioration of bone tissue. Application of pulsed electromagnetic fields (PEMF), a non-invasive method with a low complication risk, is known to stimulate bone formation. The present study examines the histomorphometric and biochemical effects of PEMF application on the healing of bone defects in rats with heparin-induced secondary osteoporosis. Briefly, 12-month-old male Sprague-Dawley rats were examined in a prospective, randomized, single-blind study. Osteoporosis was induced by administering a daily dose of 2 IU/g heparin for 33 days. Bone defects were created on the right femur on Day 35. PEMF of an average intensity of 0.8 ± 0.2 mT and a frequency of 7.3 Hz, was applied for 1 h/day, for 28 days following surgery. Bone healing was evaluated by histomorphometric and biochemical analyses. The heparin + PEMF group displayed the largest amount of new bone area (P = .002) and the lowest mean CTx on Day 63 (P = .05). This study demonstrates that heparin administration leads to bone loss and osteoporosis, whereas the application of PEMF decreases this effect.
Collapse
Affiliation(s)
- Olgun Topal
- Faculty of Dentistry, Department of Oral Maxillofacial Surgery, Afyonkarahisar Health Sciences University , Afyonkarahisar, Turkey
| | - Müge Çina Aksoy
- Faculty of Dentistry, Department of Oral Maxillofacial Surgery, Süleyman Demirel University , Isparta, Turkey
| | - İbrahim Metin Ciriş
- Faculty of Medicine, Department of Medical Pathology, Süleyman Demirel University , Isparta, Turkey
| | - Duygu Kumbul Doğuç
- Faculty of Medicine, Department of Medical Biochemistry, Süleyman Demirel University , Isparta, Turkey
| | - Seden Sert
- Faculty of Medicine, Department of Medical Biochemistry, Süleyman Demirel University , Isparta, Turkey
| | - Selçuk Çömlekçi
- Faculty of Engineering,Department of Electronics and CommunicationEngineering, Süleyman Demirel University , Isparta, Turkey
| |
Collapse
|
20
|
Leppik L, Oliveira KMC, Bhavsar MB, Barker JH. Electrical stimulation in bone tissue engineering treatments. Eur J Trauma Emerg Surg 2020; 46:231-244. [PMID: 32078704 PMCID: PMC7113220 DOI: 10.1007/s00068-020-01324-1] [Citation(s) in RCA: 129] [Impact Index Per Article: 25.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Accepted: 02/04/2020] [Indexed: 12/20/2022]
Abstract
Electrical stimulation (EStim) has been shown to promote bone healing and regeneration both in animal experiments and clinical treatments. Therefore, incorporating EStim into promising new bone tissue engineering (BTE) therapies is a logical next step. The goal of current BTE research is to develop combinations of cells, scaffolds, and chemical and physical stimuli that optimize treatment outcomes. Recent studies demonstrating EStim's positive osteogenic effects at the cellular and molecular level provide intriguing clues to the underlying mechanisms by which it promotes bone healing. In this review, we discuss results of recent in vitro and in vivo research focused on using EStim to promote bone healing and regeneration and consider possible strategies for its application to improve outcomes in BTE treatments. Technical aspects of exposing cells and tissues to EStim in in vitro and in vivo model systems are also discussed.
Collapse
Affiliation(s)
- Liudmila Leppik
- Frankfurt Initiative for Regenerative Medicine, Experimental Orthopedics and Trauma Surgery, J.W. Goethe University, Frankfurt/Main, Germany.
| | - Karla Mychellyne Costa Oliveira
- Frankfurt Initiative for Regenerative Medicine, Experimental Orthopedics and Trauma Surgery, J.W. Goethe University, Frankfurt/Main, Germany
| | - Mit Balvantray Bhavsar
- Frankfurt Initiative for Regenerative Medicine, Experimental Orthopedics and Trauma Surgery, J.W. Goethe University, Frankfurt/Main, Germany
| | - John Howard Barker
- Frankfurt Initiative for Regenerative Medicine, Experimental Orthopedics and Trauma Surgery, J.W. Goethe University, Frankfurt/Main, Germany
| |
Collapse
|
21
|
Nam S, Kim S, Yoon K, Hong HS, Son Y. Preconditioning with repetitive electromagnetic stimulation enhances activity of bone marrow mesenchymal stem cells from elderly patients through Erk1/2 via nitric oxide. Int J Mol Med 2020; 45:678-686. [PMID: 31894263 DOI: 10.3892/ijmm.2019.4450] [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: 09/30/2019] [Accepted: 12/04/2019] [Indexed: 11/06/2022] Open
Abstract
Use of bone marrow aspirate (BMA) is a clinically advantageous cell therapeutic that bypasses the need for elaborate ex vivo cell culturing. However, a low level of bone marrow‑mesenchymal stem cells (BM‑MSCs) in the BMA and weak survival rate of these cells post‑transplantation entails an insufficient efficacy in vivo. Moreover, stem cell activity in BMA is impaired by age or background diseases. Thus, in order to enrich the BM‑MSC pool and improve cell survival, novel cell preconditioning technologies are required. In this study, it has been revealed that the pretreatment of repetitive electromagnetic stimulation (rEMS) is capable of enhancing fibroblastic colony‑forming units and cell proliferation in the BM‑MSCs, possibly via transient nitric oxide production and extracellular signal regulated kinase 1/2 activation. Notably, this effect was more apparent in stem cells isolated from older patients than from young patients. Furthermore, the rEMS‑pretreated cells showed ~53% higher cell survival, compared with the untreated cells, after cell transplantation in mice with no signs of tumorigenesis. Collectively, transient rEMS preconditioning could be utilized to enhance the activity of stem cells and thus, application of rEMS preconditioning to stem cells isolated from older patients is expected to improve the therapeutic effect of stem cells.
Collapse
Affiliation(s)
- Seungwoo Nam
- Department of Genetic Engineering, College of Life Science and Graduate School of Biotechnology, Kyung Hee University, Yongin, Gyeonggi‑do 446‑701, Republic of Korea
| | - Suna Kim
- Department of Genetic Engineering, College of Life Science and Graduate School of Biotechnology, Kyung Hee University, Yongin, Gyeonggi‑do 446‑701, Republic of Korea
| | - Kangjun Yoon
- St. Peter's Hospital, Seoul 135‑859, Republic of Korea
| | - Hyun Sook Hong
- Department of Biomedical Science and Technology, Graduate School, Kyung Hee University, Republic of Korea
| | - Youngsook Son
- Department of Genetic Engineering, College of Life Science and Graduate School of Biotechnology, Kyung Hee University, Yongin, Gyeonggi‑do 446‑701, Republic of Korea
| |
Collapse
|
22
|
Cruciani S, Garroni G, Ventura C, Danani A, Nečas A, Maioli M. Stem cells and physical energies: can we really drive stem cell fate? Physiol Res 2019; 68:S375-S384. [PMID: 32118467 DOI: 10.33549/physiolres.934388] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Adult stem cells are undifferentiated elements able to self-renew or differentiate to maintain tissue integrity. Within this context, stem cells are able to divide in a symmetric fashion, feature characterising all the somatic cells, or in an asymmetric way, which leads daughter cells to different fates. It is worth highlighting that cell polarity have a critical role in regulating stem cell asymmetric division and the proper control of cell division depends on different proteins involved in cell development, differentiation and maintenance of tissue homeostasis. Moreover, the interaction between cells and the extracellular matrix are crucial in influencing cell behavior, included in terms of mechanical properties as cytoskeleton plasticity and remodelling, and membrane tension. Finally, the activation of specific transcriptional program and epigenetic modifications contributes to cell fate determination, through modulation of cellular signalling cascades. It is well known that physical and mechanical stimuli are able to influence biological systems, and in this context, the effects of electromagnetic fields (EMFs) have already shown a considerable role, even though there is a lack of knowledge and much remains to be done around this topic. In this review, we summarize the historical background of EMFs applications and the main molecular mechanism involved in cellular remodelling, with particular attention to cytoskeleton elasticity and cell polarity, required for driving stem cell behavior.
Collapse
Affiliation(s)
- S Cruciani
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy.
| | | | | | | | | | | |
Collapse
|
23
|
Implantable electrical stimulation bioreactor with liquid crystal polymer-based electrodes for enhanced bone regeneration at mandibular large defects in rabbit. Med Biol Eng Comput 2019; 58:383-399. [PMID: 31853774 DOI: 10.1007/s11517-019-02046-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2018] [Accepted: 09/05/2019] [Indexed: 02/07/2023]
Abstract
The osseous regeneration of large bone defects is still a major clinical challenge in maxillofacial and orthopedic surgery. Previous studies demonstrated that biphasic electrical stimulation (ES) stimulates bone formation; however, polyimide electrode should be removed after regeneration. This study presents an implantable electrical stimulation bioreactor with electrodes based on liquid crystal polymer (LCP), which can be permanently implanted due to excellent biocompatibility to bone tissue. The bioreactor was implanted into a critical-sized bone defect and subjected to ES for one week, where bone regeneration was evaluated four weeks after surgery using micro-CT. The effect of ES via the bioreactor was compared with a sham control group and a positive control group that received recombinant human bone morphogenetic protein (rhBMP)-2 (20 μg). New bone volume per tissue volume (BV/TV) in the ES and rhBMP-2 groups increased to 132% (p < 0.05) and 174% (p < 0.01), respectively, compared to that in the sham control group. In the histological evaluation, there was no inflammation within the bone defects and adjacent to LCP in all the groups. This study showed that the ES bioreactor with LCP electrodes could enhance bone regeneration at large bone defects, where LCP can act as a mechanically resistant outer box without inflammation. Graphical abstract To enhance bone regeneration, a bioreactor comprising collagen sponge and liquid crystal polymer-based electrode was implanted in the bone defect. Within the defect, electrical current pulses having biphasic waveform were applied from the implanted bioreactor.
Collapse
|
24
|
Bernardo R, Rodrigues A, Soares Dos Santos MP, Carneiro P, Lopes A, Sequeira Amaral J, Sequeira Amaral V, Morais R. Novel magnetic stimulation methodology for low-current implantable medical devices. Med Eng Phys 2019; 73:77-84. [PMID: 31477429 DOI: 10.1016/j.medengphy.2019.07.015] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Revised: 05/10/2019] [Accepted: 07/28/2019] [Indexed: 12/15/2022]
Abstract
Recent studies highlight the ability of inductive architectures to deliver therapeutic magnetic stimuli to target tissues and to be embedded into small-scale intracorporeal medical devices. However, to date, current micro-scale biomagnetic devices require very high electric current excitations (usually exceeding 1 A) to ensure the delivery of efficient magnetic flux densities. This is a critical problem as advanced implantable devices demand self-powering, stand-alone and long-term operation. This work provides, for the first time, a novel small-scale magnetic stimulation system that requires up to 50-fold lower electric current excitations than required by relevant biomagnetic technology recently proposed. Computational models were developed to analyse the magnetic stimuli distributions and densities delivered to cellular tissues during in vitro experiments, such that the feasibility of this novel stimulator can be firstly evaluated on cell culture tests. The results demonstrate that this new stimulative technology is able to deliver osteogenic stimuli (0.1-7 mT range) by current excitations in the 0.06-4.3 mA range. Moreover, it allows coil designs with heights lower than 1 mm without significant loss of magnetic stimuli capability. Finally, suitable core diameters and stimulator-stimulator distances allow to define heterogeneity or quasi-homogeneity stimuli distributions. These results support the design of high-sophisticated biomagnetic devices for a wide range of therapeutic applications.
Collapse
Affiliation(s)
- Rodrigo Bernardo
- Department of Mechanical Engineering, University of Aveiro, Aveiro, Portugal
| | - André Rodrigues
- Department of Mechanical Engineering, University of Aveiro, Aveiro, Portugal
| | - Marco P Soares Dos Santos
- Department of Mechanical Engineering, University of Aveiro, Aveiro, Portugal; Centre for Mechanical Technology and Automation (TEMA), University of Aveiro, Aveiro, Portugal; Associated Laboratory for Energy, Transports and Aeronautics (LAETA), Portugal.
| | - Pedro Carneiro
- Department of Mechanical Engineering, University of Aveiro, Aveiro, Portugal
| | - António Lopes
- Department of Physics, University of Aveiro, Aveiro, Portugal; Aveiro Institute of Materials, Aveiro, Portugal
| | - João Sequeira Amaral
- Department of Physics, University of Aveiro, Aveiro, Portugal; Aveiro Institute of Materials, Aveiro, Portugal
| | - Vítor Sequeira Amaral
- Department of Physics, University of Aveiro, Aveiro, Portugal; Aveiro Institute of Materials, Aveiro, Portugal
| | - Raul Morais
- University of Trás-os-Montes e Alto Douro, Vila Real, Portugal; Institute for Systems and Computer Engineering, Technology and Science (INESC TEC), Porto, Portugal
| |
Collapse
|
25
|
Wang H, Tang X, Li W, Chen J, Li H, Yan J, Yuan X, Wu H, Liu C. Enhanced osteogenesis of bone marrow stem cells cultured on hydroxyapatite/collagen I scaffold in the presence of low-frequency magnetic field. JOURNAL OF MATERIALS SCIENCE. MATERIALS IN MEDICINE 2019; 30:89. [PMID: 31342178 DOI: 10.1007/s10856-019-6289-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2018] [Accepted: 01/29/2019] [Indexed: 06/10/2023]
Abstract
As a non-invasive biophysical therapy, electromagnetic fields (EMF) have been widely used to promote the healing of fractures. In the present study, hydroxyapatite/collagen I (HAC) loaded with rabbit bone marrow mesenchymal stem cells (MSCs) were cultured in a dynamic perfusion bioreactor and exposed to EMF of 15 Hz/1mT. Osteogenic differentiation of the seeded cells was analyzed through the evaluation of ALP activity and osteogenesis-related genes expression in vitro. The in vivo osteogenesis efficacy of the cell laden HAC constructs treated with/without EMF was evaluated through a rabbit femur condyle defect model. The results showed that EMF of 15 Hz/1mT could enhance the osteogenic differentiation of the cells seeded on HAC scaffold. Furthermore, the in vivo experiments demonstrated that EMF exposure could promote bone regeneration within the defect and bone integration between the graft and host bone. Taking together, the MSCs seeded HAC scaffold combined with EMF exposure could be a promising approach for bone tissue engineering.
Collapse
Affiliation(s)
- Huaixi Wang
- Department of Spine and Spinal Cord Surgery, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, No. 7 Weiwu road, 450003, Zhengzhou, P. R. China
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Avenue 1095, 430030, Wuhan, P. R. China
| | - Xiangyu Tang
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Avenue 1095, 430030, Wuhan, P. R. China
| | - Wenkai Li
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Avenue 1095, 430030, Wuhan, P. R. China
| | - Jingyuan Chen
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Avenue 1095, 430030, Wuhan, P. R. China
| | - Hao Li
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Avenue 1095, 430030, Wuhan, P. R. China
| | - Jiyuan Yan
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Avenue 1095, 430030, Wuhan, P. R. China
| | - Xuefeng Yuan
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Avenue 1095, 430030, Wuhan, P. R. China
| | - Hua Wu
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Avenue 1095, 430030, Wuhan, P. R. China.
| | - Chaoxu Liu
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Avenue 1095, 430030, Wuhan, P. R. China.
| |
Collapse
|
26
|
Miyamoto H, Sawaji Y, Iwaki T, Masaoka T, Fukada E, Date M, Yamamoto K. Intermittent pulsed electromagnetic field stimulation activates the mTOR pathway and stimulates the proliferation of osteoblast‐like cells. Bioelectromagnetics 2019; 40:412-421. [DOI: 10.1002/bem.22207] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Accepted: 06/26/2019] [Indexed: 02/04/2023]
Affiliation(s)
- Hironori Miyamoto
- Department of Orthopedic SurgeryTokyo Medical University Tokyo Japan
| | - Yasunobu Sawaji
- Department of Orthopedic SurgeryTokyo Medical University Tokyo Japan
| | - Takahiro Iwaki
- Department of Orthopedic SurgeryTokyo Medical University Tokyo Japan
| | - Toshinori Masaoka
- Department of Orthopedic SurgeryTokyo Medical University Tokyo Japan
| | - Eiichi Fukada
- Laboratory of PiezoelectricityKobayasi Institute of Physical Research Tokyo Japan
| | - Munehiro Date
- Laboratory of PiezoelectricityKobayasi Institute of Physical Research Tokyo Japan
| | - Kengo Yamamoto
- Department of Orthopedic SurgeryTokyo Medical University Tokyo Japan
| |
Collapse
|
27
|
Costantini E, Sinjari B, D'Angelo C, Murmura G, Reale M, Caputi S. Human Gingival Fibroblasts Exposed to Extremely Low-Frequency Electromagnetic Fields: In Vitro Model of Wound-Healing Improvement. Int J Mol Sci 2019; 20:ijms20092108. [PMID: 31035654 PMCID: PMC6540598 DOI: 10.3390/ijms20092108] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Revised: 03/23/2019] [Accepted: 04/25/2019] [Indexed: 01/24/2023] Open
Abstract
Several clinical studies have suggested the impact of sinusoidal and pulsed electromagnetic fields in quickening wound repair processes and tissue regeneration. The clinical use of extremely low-frequency electromagnetic fields could represent a novel frontier in tissue repair and oral health, with an interesting clinical perspective. The present study aimed to evaluate the effect of an extremely low-frequency sinusoidal electromagnetic field (SEMF) and an extremely low-frequency pulsed electromagnetic field (PEMF) with flux densities of 1 mT on a model of oral healing process using gingival fibroblasts. An in vitro mechanical injury was produced to evaluate wound healing, migration, viability, metabolism, and the expression of selected cytokines and protease genes in fibroblasts exposed to or not exposed to the SEMF and the PEMF. Interleukin 6 (IL-6), transforming growth factor beta 1 (TGF-β), metalloproteinase 2 (MMP-2), monocyte chemoattractant protein 1 (MCP-1), inducible nitric oxide synthase (iNOS), and heme oxygenase 1 (HO-1) are involved in wound healing and tissue regeneration, favoring fibroblast proliferation, chemotaxis, and activation. Our results show that the exposure to each type of electromagnetic field increases the early expression of IL-6, TGF-β, and iNOS, driving a shift from an inflammatory to a proliferative phase of wound repair. Additionally, a later induction of MMP-2, MCP-1, and HO-1 was observed after electromagnetic field exposure, which quickened the wound-healing process. Moreover, electromagnetic field exposure influenced the proliferation, migration, and metabolism of human gingival fibroblasts compared to sham-exposed cells. This study suggests that exposure to SEMF and PEMF could be an interesting new non-invasive treatment option for wound healing. However, additional studies are needed to elucidate the best exposure conditions to provide the desired in vivo treatment efficacy.
Collapse
Affiliation(s)
- Erica Costantini
- Department of Medical, Oral and Biotechnological Science, University "G. d'Annunzio" Chieti-Pescara, 66100 Chieti, Italy.
| | - Bruna Sinjari
- Department of Medical, Oral and Biotechnological Science, University "G. d'Annunzio" Chieti-Pescara, 66100 Chieti, Italy.
| | - Chiara D'Angelo
- Department of Medical, Oral and Biotechnological Science, University "G. d'Annunzio" Chieti-Pescara, 66100 Chieti, Italy.
| | - Giovanna Murmura
- Department of Medical, Oral and Biotechnological Science, University "G. d'Annunzio" Chieti-Pescara, 66100 Chieti, Italy.
| | - Marcella Reale
- Department of Medical, Oral and Biotechnological Science, University "G. d'Annunzio" Chieti-Pescara, 66100 Chieti, Italy.
| | - Sergio Caputi
- Department of Medical, Oral and Biotechnological Science, University "G. d'Annunzio" Chieti-Pescara, 66100 Chieti, Italy.
| |
Collapse
|
28
|
Galli C, Pedrazzi G, Guizzardi S. The cellular effects of Pulsed Electromagnetic Fields on osteoblasts: A review. Bioelectromagnetics 2019; 40:211-233. [PMID: 30908726 DOI: 10.1002/bem.22187] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Accepted: 03/08/2019] [Indexed: 12/12/2022]
Abstract
Electromagnetic fields (EMFs) have long been known to interact with living organisms and their cells and to bear the potential for therapeutic use. Among the most extensively investigated applications, the use of Pulsed EMFs (PEMFs) has proven effective to ameliorate bone healing in several studies, although the evidence is still inconclusive. This is due in part to our still-poor understanding of the mechanisms by which PEMFs act on cells and affect their functions and to an ongoing lack of consensus on the most effective parameters for specific clinical applications. The present review has compared in vitro studies on PEMFs on different osteoblast models, which elucidate potential mechanisms of action for PEMFs, up to the most recent insights into the role of primary cilia, and highlight the critical issues underlying at least some of the inconsistent results in the available literature. Bioelectromagnetics. 2019;9999:XX-XX. © 2019 Bioelectromagnetics Society.
Collapse
Affiliation(s)
- Carlo Galli
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Giuseppe Pedrazzi
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Stefano Guizzardi
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| |
Collapse
|
29
|
Wang T, Yang L, Jiang J, Liu Y, Fan Z, Zhong C, He C. Pulsed electromagnetic fields: promising treatment for osteoporosis. Osteoporos Int 2019; 30:267-276. [PMID: 30603841 DOI: 10.1007/s00198-018-04822-6] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Accepted: 12/18/2018] [Indexed: 02/05/2023]
Abstract
Osteoporosis (OP) is considered to be a well-defined disease which results in high morbidity and mortality. In patients diagnosed with OP, low bone mass and fragile bone strength have been demonstrated to significantly increase risk of fragility fractures. To date, various anabolic and antiresorptive therapies have been applied to maintain healthy bone mass and strength. Pulsed electromagnetic fields (PEMFs) are employed to treat patients suffering from delayed fracture healing and nonunions. Although PEMFs stimulate osteoblastogenesis, suppress osteoclastogenesis, and influence the activity of bone marrow mesenchymal stem cells (BMSCs) and osteocytes, ultimately leading to retention of bone mass and strength. However, whether PEMFs could be taken into clinical use to treat OP is still unknown. Furthermore, the deeper signaling pathways underlying the way in which PEMFs influence OP remain unclear.
Collapse
Affiliation(s)
- T Wang
- Department of Rehabilitation Medicine, West China Hospital, Sichuan University, No. 37 Guoxue Xiang, Chengdu, 610041, Sichuan, People's Republic of China
- Key Laboratory of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, People's Republic of China
| | - L Yang
- Department of Rehabilitation Medicine, West China Hospital, Sichuan University, No. 37 Guoxue Xiang, Chengdu, 610041, Sichuan, People's Republic of China
- Key Laboratory of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, People's Republic of China
| | - J Jiang
- Department of Rehabilitation Medicine, West China Hospital, Sichuan University, No. 37 Guoxue Xiang, Chengdu, 610041, Sichuan, People's Republic of China
- Key Laboratory of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, People's Republic of China
| | - Y Liu
- Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Z Fan
- Department of Rehabilitation Medicine, West China Hospital, Sichuan University, No. 37 Guoxue Xiang, Chengdu, 610041, Sichuan, People's Republic of China
- Key Laboratory of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, People's Republic of China
| | - C Zhong
- Department of Rehabilitation Medicine, West China Hospital, Sichuan University, No. 37 Guoxue Xiang, Chengdu, 610041, Sichuan, People's Republic of China
- Key Laboratory of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, People's Republic of China
| | - C He
- Department of Rehabilitation Medicine, West China Hospital, Sichuan University, No. 37 Guoxue Xiang, Chengdu, 610041, Sichuan, People's Republic of China.
- Key Laboratory of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, People's Republic of China.
| |
Collapse
|
30
|
The Use of Pulsed Electromagnetic Fields to Promote Bone Responses to Biomaterials In Vitro and In Vivo. Int J Biomater 2018; 2018:8935750. [PMID: 30254677 PMCID: PMC6140132 DOI: 10.1155/2018/8935750] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Accepted: 08/09/2018] [Indexed: 12/14/2022] Open
Abstract
Implantable biomaterials are extensively used to promote bone regeneration or support endosseous prosthesis in orthopedics and dentistry. Their use, however, would benefit from additional strategies to improve bone responses. Pulsed Electromagnetic Fields (PEMFs) have long been known to act on osteoblasts and bone, affecting their metabolism, in spite of our poor understanding of the underlying mechanisms. Hence, we have the hypothesis that PEMFs may also ameliorate cell responses to biomaterials, improving their growth, differentiation, and the expression of a mature phenotype and therefore increasing the tissue integration of the implanted devices and their clinical success. A broad range of settings used for PEMFs stimulation still represents a hurdle to better define treatment protocols and extensive research is needed to overcome this issue. The present review includes studies that investigated the effects of PEMFs on the response of bone cells to different classes of biomaterials and the reports that focused on in vivo investigations of biomaterials implanted in bone.
Collapse
|
31
|
Osteogenic Effect and Cell Signaling Activation of Extremely Low-Frequency Pulsed Electromagnetic Fields in Adipose-Derived Mesenchymal Stromal Cells. Stem Cells Int 2018; 2018:5402853. [PMID: 30123287 PMCID: PMC6079332 DOI: 10.1155/2018/5402853] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Accepted: 06/06/2018] [Indexed: 11/27/2022] Open
Abstract
Extremely low-frequency pulsed electromagnetic field (ELF-PEMF) devices have been used in the clinic for the treatment of bone disorders over the past 30 years. However, the underlying mechanism of which ELF-PEMFs exert an effect on tissues at a cellular level is not well understood. Hence, in this study, we explored the potential of different ELF-PEMF signals in modulating human adipose-derived mesenchymal stromal cells' (hAMSC) osteogenic capability. The cell proliferation rate was assessed using carboxyfluorescein succinimidyl ester (CFSE) method. The osteogenesis potential of cells was determined by alkaline phosphatase (ALP) activity, Alizarin-Red S staining, and RT-qPCR. Finally, the intracellular signaling pathway of a selected ELF-PEMF signal was examined using the PathScan Intracellular Signaling Array. Among the tested ELF-PEMF signals, program 20 (26 Hz) showed activation of the Akt and MAPK/ERK signaling cascade and significant upregulations of collagen I, alkaline phosphatase, and osteocalcin when compared to nonstimulated cells. This study demonstrates the potential of certain ELF-PEMF signal parameters to induce osteogenic differentiation of hAMSC and provides important clues in terms of the molecular mechanisms for the stimulation of osteogenic effects by ELF-PEMF on hAMSC.
Collapse
|
32
|
Bloise N, Petecchia L, Ceccarelli G, Fassina L, Usai C, Bertoglio F, Balli M, Vassalli M, Cusella De Angelis MG, Gavazzo P, Imbriani M, Visai L. The effect of pulsed electromagnetic field exposure on osteoinduction of human mesenchymal stem cells cultured on nano-TiO2 surfaces. PLoS One 2018; 13:e0199046. [PMID: 29902240 PMCID: PMC6002089 DOI: 10.1371/journal.pone.0199046] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Accepted: 05/30/2018] [Indexed: 12/11/2022] Open
Abstract
Human bone marrow-derived mesenchymal stem cells (hBM-MSCs) are considered a great promise in the repair and regeneration of bone. Considerable efforts have been oriented towards uncovering the best strategy to promote stem cells osteogenic differentiation. In previous studies, hBM-MSCs exposed to physical stimuli such as pulsed electromagnetic fields (PEMFs) or directly seeded on nanostructured titanium surfaces (TiO2) were shown to improve their differentiation to osteoblasts in osteogenic condition. In the present study, the effect of a daily PEMF-exposure on osteogenic differentiation of hBM-MSCs seeded onto nanostructured TiO2 (with clusters under 100 nm of dimension) was investigated. TiO2-seeded cells were exposed to PEMF (magnetic field intensity: 2 mT; intensity of induced electric field: 5 mV; frequency: 75 Hz) and examined in terms of cell physiology modifications and osteogenic differentiation. Results showed that PEMF exposure affected TiO2-seeded cells osteogenesis by interfering with selective calcium-related osteogenic pathways, and greatly enhanced hBM-MSCs osteogenic features such as the expression of early/late osteogenic genes and protein production (e.g., ALP, COL-I, osteocalcin and osteopontin) and ALP activity. Finally, PEMF-treated cells resulted to secrete into conditioned media higher amounts of BMP-2, DCN and COL-I than untreated cell cultures. These findings confirm once more the osteoinductive potential of PEMF, suggesting that its combination with TiO2 nanostructured surface might be a great option in bone tissue engineering applications.
Collapse
Affiliation(s)
- Nora Bloise
- Department of Molecular Medicine (DMM), Centre for Health Technologies (C.H.T.), INSTM Unit, University of Pavia, Pavia, Italy
- Department of Occupational Medicine, Toxicology and Environmental Risks, Istituti Clinici Scientifici Maugeri, IRCCS, Pavia, Italy
- * E-mail: (NB); (LV)
| | | | - Gabriele Ceccarelli
- Department of Public Health, Experimental Medicine and Forensic, Centre for Health Technologies (C.H.T.), Human Anatomy Unit, University of Pavia, Pavia, Italy
| | - Lorenzo Fassina
- Department of Electrical, Computer and Biomedical Engineering, Centre for Health Technologies (C.H.T.), University of Pavia, Pavia, Italy
| | - Cesare Usai
- Institute of Biophysics, National Research Council, Genova, Italy
| | - Federico Bertoglio
- Department of Molecular Medicine (DMM), Centre for Health Technologies (C.H.T.), INSTM Unit, University of Pavia, Pavia, Italy
- Department of Occupational Medicine, Toxicology and Environmental Risks, Istituti Clinici Scientifici Maugeri, IRCCS, Pavia, Italy
| | - Martina Balli
- Department of Public Health, Experimental Medicine and Forensic, Centre for Health Technologies (C.H.T.), Human Anatomy Unit, University of Pavia, Pavia, Italy
| | - Massimo Vassalli
- Institute of Biophysics, National Research Council, Genova, Italy
| | - Maria Gabriella Cusella De Angelis
- Department of Public Health, Experimental Medicine and Forensic, Centre for Health Technologies (C.H.T.), Human Anatomy Unit, University of Pavia, Pavia, Italy
| | - Paola Gavazzo
- Institute of Biophysics, National Research Council, Genova, Italy
| | - Marcello Imbriani
- Department of Occupational Medicine, Toxicology and Environmental Risks, Istituti Clinici Scientifici Maugeri, IRCCS, Pavia, Italy
- Department of Public Health, Experimental Medicine and Forensic, Centre for Health Technologies (C.H.T.), Human Anatomy Unit, University of Pavia, Pavia, Italy
| | - Livia Visai
- Department of Molecular Medicine (DMM), Centre for Health Technologies (C.H.T.), INSTM Unit, University of Pavia, Pavia, Italy
- Department of Occupational Medicine, Toxicology and Environmental Risks, Istituti Clinici Scientifici Maugeri, IRCCS, Pavia, Italy
- * E-mail: (NB); (LV)
| |
Collapse
|
33
|
Shuai C, Yang W, Peng S, Gao C, Guo W, Lai Y, Feng P. Physical stimulations and their osteogenesis-inducing mechanisms. Int J Bioprint 2018; 4:138. [PMID: 33102916 PMCID: PMC7581999 DOI: 10.18063/ijb.v4i2.138] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Accepted: 05/09/2018] [Indexed: 12/27/2022] Open
Abstract
Physical stimulations such as magnetic, electric and mechanical stimulation could enhance cell activity and promote bone formation in bone repair process via activating signal pathways, modulating ion channels, regulating bonerelated gene expressions, etc. In this paper, bioeffects of physical stimulations on cell activity, tissue growth and bone healing were systematically summarized, which especially focused on their osteogenesis-inducing mechanisms. Detailedly, magnetic stimulation could produce Hall effect which improved the permeability of cell membrane and promoted the migration of ions, especially accelerating the extracellular calcium ions to pass through cell membrane. Electric stimulation could induce inverse piezoelectric effect which generated electric signals, accordingly up-regulating intracellular calcium levels and growth factor synthesis. And mechanical stimulation could produce mechanical signals which were converted into corresponding biochemical signals, thus activating various signaling pathways on cell membrane and inducing a series of gene expressions. Besides, bioeffects of physical stimulations combined with bone scaffolds which fabricated using 3D printing technology on bone cells were discussed. The equipments of physical stimulation system were described. The opportunities and challenges of physical stimulations were also presented from the perspective of bone repair.
Collapse
Affiliation(s)
- Cijun Shuai
- State Key Laboratory of High Performance Complex Manufacturing, College of Mechanical and Electrical Engineering, Central South University, Changsha, China.,Jiangxi University of Science and Technology, Ganzhou, China.,Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Changsha, China
| | - Wenjing Yang
- State Key Laboratory of High Performance Complex Manufacturing, College of Mechanical and Electrical Engineering, Central South University, Changsha, China
| | - Shuping Peng
- Hunan Provincial Tumor Hospital and the Affiliated Tumor Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Chengde Gao
- State Key Laboratory of High Performance Complex Manufacturing, College of Mechanical and Electrical Engineering, Central South University, Changsha, China
| | - Wang Guo
- State Key Laboratory of High Performance Complex Manufacturing, College of Mechanical and Electrical Engineering, Central South University, Changsha, China
| | - Yuxiao Lai
- Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, China
| | - Pei Feng
- State Key Laboratory of High Performance Complex Manufacturing, College of Mechanical and Electrical Engineering, Central South University, Changsha, China
| |
Collapse
|
34
|
Lalegül-Ülker Ö, Elçin AE, Elçin YM. Intrinsically Conductive Polymer Nanocomposites for Cellular Applications. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1078:135-153. [PMID: 30357622 DOI: 10.1007/978-981-13-0950-2_8] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Intrinsically conductive polymer nanocomposites have a remarkable potential for cellular applications such as biosensors, drug delivery systems, cell culture systems and tissue engineering biomaterials. Intrinsically conductive polymers transmit electrical stimuli between cells, and induce regeneration of electroactive tissues such as muscle, nerve, bone and heart. However, biocompatibility and processability are common issues for intrinsically conductive polymers. Conductive polymer composites are gaining importance for tissue engineering applications due to their excellent mechanical, electrical, optical and chemical functionalities. Here, we summarize the different types of intrinsically conductive polymers containing electroactive nanocomposite systems. Cellular applications of conductive polymer nanocomposites are also discussed focusing mainly on poly(aniline), poly(pyrrole), poly(3,4-ethylene dioxythiophene) and poly(thiophene).
Collapse
Affiliation(s)
- Özge Lalegül-Ülker
- Tissue Engineering, Biomaterials and Nanobiotechnology Laboratory, Faculty of Science, Stem Cell Institute, Ankara University, Ankara, Turkey
| | - Ayşe Eser Elçin
- Tissue Engineering, Biomaterials and Nanobiotechnology Laboratory, Faculty of Science, Stem Cell Institute, Ankara University, Ankara, Turkey
| | - Yaşar Murat Elçin
- Tissue Engineering, Biomaterials and Nanobiotechnology Laboratory, Faculty of Science, Stem Cell Institute, Ankara University, Ankara, Turkey. .,Biovalda Health Technologies, Inc., Ankara, Turkey.
| |
Collapse
|
35
|
Unsdorfer KML, Abdelmagid SM. Percutaneous Kirschner Wire Repair of Failed Digital Arthrodesis Using Pulsed Electromagnetic Field Therapy A Case Report. J Am Podiatr Med Assoc 2018; 108:77-80. [PMID: 29547026 DOI: 10.7547/16-007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
A case of a repaired failed toe arthrodesis is presented. A novel and previously unreported approach using a percutaneously placed Kirschner wire coupled with a pulsed electromagnetic field achieved healing of a painful pseudoarthrosis at 54 days. With a percutaneous technique, open debridement of the failed arthrodesis site can be avoided.
Collapse
Affiliation(s)
| | - Samir M. Abdelmagid
- Department of Anatomy and Neurobiology, Northeast Ohio Medical University, Rootstown, OH
| |
Collapse
|
36
|
Yang X, He H, Gao Q, He C. Pulsed electromagnetic field improves subchondral bone microstructure in knee osteoarthritis rats through a Wnt/β-catenin signaling-associated mechanism. Bioelectromagnetics 2017; 39:89-97. [PMID: 29251361 DOI: 10.1002/bem.22106] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2017] [Accepted: 11/22/2017] [Indexed: 02/05/2023]
Affiliation(s)
- Xiaotian Yang
- Department of Rehabilitation Medicine; West China Hospital; Sichuan University; Chengdu China
- Key Laboratory of Rehabilitation Medicine in Sichuan; Chengdu China
| | - Hongchen He
- Department of Rehabilitation Medicine; West China Hospital; Sichuan University; Chengdu China
- Key Laboratory of Rehabilitation Medicine in Sichuan; Chengdu China
| | - Qiang Gao
- Department of Rehabilitation Medicine; West China Hospital; Sichuan University; Chengdu China
- Key Laboratory of Rehabilitation Medicine in Sichuan; Chengdu China
| | - Chengqi He
- Department of Rehabilitation Medicine; West China Hospital; Sichuan University; Chengdu China
- Key Laboratory of Rehabilitation Medicine in Sichuan; Chengdu China
| |
Collapse
|
37
|
Pulsed magnetic field treatment as antineuropathic pain therapy. Rev Neurosci 2017; 28:751-758. [DOI: 10.1515/revneuro-2017-0003] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2017] [Accepted: 03/08/2017] [Indexed: 01/08/2023]
Abstract
AbstractNo satisfactory effective therapy is still available to treat trauma- or disease-induced neuropathic pain, and current available treatment options have several side effects. Pulsed magnetic field (PMF) treatments are receiving growing interest as a therapeutic approach for several neuronal diseases. Although the exact mechanism of action of PMF treatments is unknown, reported findings represent a promising alternative therapeutic choice for the management of neuropathic pain. PMF treatments can supply new strategies for the therapy of life-threatening neuropathic pain due to its antihyperglycemic, anti-inflammatory, antihyperalgesic, antiallodynic, and neuroimmunomodulatory actions. In this review, I summarized the several recent findings about antineuropathic actions of PMF treatment in experimental animals with neuropathic pain induced by disease and/or damage.
Collapse
|
38
|
Effects of low-intensity pulsed electromagnetic fields on bone microarchitecture, mechanical strength and bone turnover in type 2 diabetic db/db mice. Sci Rep 2017; 7:10834. [PMID: 28883516 PMCID: PMC5589741 DOI: 10.1038/s41598-017-11090-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Accepted: 08/17/2017] [Indexed: 01/06/2023] Open
Abstract
Type 2 diabetic patients have impaired bone quality, leading to increased fracture risk. Substantial evidence demonstrates that pulsed electromagnetic fields (PEMF) could resist osteopenia/osteoporosis induced by estrogen deficiency and disuse. However, the effects of PEMF on osteopenia/osteoporosis associated with diabetes, especially for more prevalent type 2 diabetes, remain poorly understood. We herein investigated the skeletal effects and mechanisms of PEMF (15 Hz, 20 Gs) on leptin receptor-deficient db/db mice with typical type 2 diabetic symptoms. Our µCT results showed that 12-week PEMF exposure significantly improved both cancellous and cortical bone microarchitecture in db/db mice. Three-point bending and biomechanical indentation testing demonstrated that PEMF improved whole-bone structural properties and tissue-level material properties in db/db mice. PEMF significantly promoted bone formation in db/db mice evidenced by increased serum osteocalcin and bone mineral apposition rate, whereas PEMF exerted no observable alteration in bone resorption. Real-time PCR showed that PEMF upregulated tibial gene expression of osteoblastogenesis-related of canonical Wnt/β-catenin signaling but not osteoclastogenesis-related RANKL-RANK signaling in db/db mice. Our findings demonstrate that PEMF improved bone quantity and quality with obvious anabolic activities in db/db mice, and imply that PEMF might become a clinically applicable treatment modality for improving bone quality in type 2 diabetic patients.
Collapse
|
39
|
Tong J, Sun L, Zhu B, Fan Y, Ma X, Yu L, Zhang J. Pulsed electromagnetic fields promote the proliferation and differentiation of osteoblasts by reinforcing intracellular calcium transients. Bioelectromagnetics 2017; 38:541-549. [DOI: 10.1002/bem.22076] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Accepted: 07/22/2017] [Indexed: 12/11/2022]
Affiliation(s)
- Jie Tong
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology; Xi'an Jiaotong University; Xi'an China
| | - Lijun Sun
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology; Xi'an Jiaotong University; Xi'an China
| | - Bin Zhu
- Xi Jing University; Xi'an China
| | - Yun Fan
- Xi Jing University; Xi'an China
| | - Xingfeng Ma
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology; Xi'an Jiaotong University; Xi'an China
| | - Liyin Yu
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology; Xi'an Jiaotong University; Xi'an China
| | - Jianbao Zhang
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology; Xi'an Jiaotong University; Xi'an China
| |
Collapse
|
40
|
Zhu S, He H, Zhang C, Wang H, Gao C, Yu X, He C. Effects of pulsed electromagnetic fields on postmenopausal osteoporosis. Bioelectromagnetics 2017; 38:406-424. [PMID: 28665487 DOI: 10.1002/bem.22065] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2016] [Accepted: 06/05/2017] [Indexed: 02/05/2023]
Abstract
Postmenopausal osteoporosis (PMOP) is considered to be a well-defined subject that has caused high morbidity and mortality. In elderly women diagnosed with PMOP, low bone mass and fragile bone strength have been proven to significantly increase risk of fragility fractures. Currently, various anabolic and anti-resorptive therapies have been employed in an attempt to retain healthy bone mass and strength. Pulsed electromagnetic fields (PEMFs), first applied in treating patients with delayed fracture healing and nonunions, may turn out to be another potential and effective therapy for PMOP. PEMFs can enhance osteoblastogenesis and inhibit osteoclastogenesis, thus contributing to an increase in bone mass and strength. However, accurate mechanisms of the positive effects of PEMFs on PMOP remain to be further elucidated. This review attempts to summarize recent advances of PEMFs in treating PMOP based on clinical trials, and animal and cellular studies. Possible mechanisms are also introduced, and the future possibility of application of PEMFs on PMOP are further explored and discussed. Bioelectromagnetics. 38:406-424, 2017. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Siyi Zhu
- Rehabilitation Medicine Center, West China Hospital, Sichuan University, Chengdu, P. R. China
- Rehabilitation Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, P. R. China
- Laboratory of Endocrinology and Metabolism, Department of Endocrinology, National Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, P. R. China
| | - Hongchen He
- Rehabilitation Medicine Center, West China Hospital, Sichuan University, Chengdu, P. R. China
| | - Chi Zhang
- Rehabilitation Medicine Center, West China Hospital, Sichuan University, Chengdu, P. R. China
- Rehabilitation Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, P. R. China
| | - Haiming Wang
- Rehabilitation Medicine Center, West China Hospital, Sichuan University, Chengdu, P. R. China
- Rehabilitation Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, P. R. China
| | - Chengfei Gao
- Rehabilitation Medicine Center, West China Hospital, Sichuan University, Chengdu, P. R. China
- Rehabilitation Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, P. R. China
| | - Xijie Yu
- Laboratory of Endocrinology and Metabolism, Department of Endocrinology, National Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, P. R. China
| | - Chengqi He
- Rehabilitation Medicine Center, West China Hospital, Sichuan University, Chengdu, P. R. China
- Rehabilitation Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, P. R. China
| |
Collapse
|
41
|
D'Amora U, Russo T, Gloria A, Rivieccio V, D'Antò V, Negri G, Ambrosio L, De Santis R. 3D additive-manufactured nanocomposite magnetic scaffolds: Effect of the application mode of a time-dependent magnetic field on hMSCs behavior. Bioact Mater 2017; 2:138-145. [PMID: 29744423 PMCID: PMC5935178 DOI: 10.1016/j.bioactmat.2017.04.003] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Revised: 04/10/2017] [Accepted: 04/12/2017] [Indexed: 01/15/2023] Open
Abstract
Over the past few years, the influence of static or dynamic magnetic fields on biological systems has become a topic of considerable interest. Magnetism has recently been implicated to play significant roles in the regulation of cell responses and, for this reason, it is revolutionizing many aspects of healthcare, also suggesting new opportunities in tissue engineering. The aim of the present study was to analyze the effect of the application mode of a time-dependent magnetic field on the behavior of human mesenchymal stem cells (hMSCs) seeded on 3D additive-manufactured poly(ɛ-caprolactone)/iron-doped hydroxyapatite (PCL/FeHA) nanocomposite scaffolds. Additive Manufacturing for bone tissue engineering. Analysis of the effect of the application mode of a time-dependent magnetic field on the cell-behavior. 3D nanocomposite magnetic scaffolds.
Collapse
Affiliation(s)
- Ugo D'Amora
- Institute of Polymers, Composites and Biomaterials, National Research Council, Naples, Italy
| | - Teresa Russo
- Institute of Polymers, Composites and Biomaterials, National Research Council, Naples, Italy
| | - Antonio Gloria
- Institute of Polymers, Composites and Biomaterials, National Research Council, Naples, Italy
| | | | - Vincenzo D'Antò
- Department of Neuroscience, Reproductive Sciences and Oral Sciences, University of Naples Federico II, Naples, Italy.,Unit of Dentistry, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | | | - Luigi Ambrosio
- Institute of Polymers, Composites and Biomaterials, National Research Council, Naples, Italy
| | - Roberto De Santis
- Institute of Polymers, Composites and Biomaterials, National Research Council, Naples, Italy
| |
Collapse
|
42
|
Yang X, He H, Zhou Y, Zhou Y, Gao Q, Wang P, He C. Pulsed electromagnetic field at different stages of knee osteoarthritis in rats induced by low-dose monosodium iodoacetate: Effect on subchondral trabecular bone microarchitecture and cartilage degradation. Bioelectromagnetics 2016; 38:227-238. [PMID: 28026095 DOI: 10.1002/bem.22028] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2016] [Accepted: 12/03/2016] [Indexed: 02/05/2023]
Affiliation(s)
- Xiaotian Yang
- Department of Rehabilitation Medicine; West China Hospital; Sichuan University; Chengdu China
- Key Laboratory of Rehabilitation Medicine in Sichuan; Chengdu China
| | - Hongchen He
- Department of Rehabilitation Medicine; West China Hospital; Sichuan University; Chengdu China
- Key Laboratory of Rehabilitation Medicine in Sichuan; Chengdu China
| | - Yuan Zhou
- Institute for Disaster Management and Reconstruction of Sichuan University and Hong Kong Polytechnic University; Chengdu China
| | - Yujing Zhou
- Department of Rehabilitation Medicine; West China Hospital; Sichuan University; Chengdu China
- Key Laboratory of Rehabilitation Medicine in Sichuan; Chengdu China
| | - Qiang Gao
- Department of Rehabilitation Medicine; West China Hospital; Sichuan University; Chengdu China
- Key Laboratory of Rehabilitation Medicine in Sichuan; Chengdu China
| | - Pu Wang
- Department of Rehabilitation Medicine; West China Hospital; Sichuan University; Chengdu China
- Key Laboratory of Rehabilitation Medicine in Sichuan; Chengdu China
| | - Chengqi He
- Department of Rehabilitation Medicine; West China Hospital; Sichuan University; Chengdu China
- Key Laboratory of Rehabilitation Medicine in Sichuan; Chengdu China
- Institute for Disaster Management and Reconstruction of Sichuan University and Hong Kong Polytechnic University; Chengdu China
| |
Collapse
|
43
|
Wang T, Wang P, Cao Z, Wang X, Wang D, Shen Y, Jing D, Luo E, Tang W. Effects of BMP9 and pulsed electromagnetic fields on the proliferation and osteogenic differentiation of human periodontal ligament stem cells. Bioelectromagnetics 2016; 38:63-77. [PMID: 27859405 DOI: 10.1002/bem.22018] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Accepted: 10/19/2016] [Indexed: 12/30/2022]
Affiliation(s)
- Tingting Wang
- Department of Stomatology; Changhai Hospital; Second Military Medical University; Shanghai China
| | - Pei Wang
- Department of Stomatology; Changhai Hospital; Second Military Medical University; Shanghai China
| | - Zhizhong Cao
- Department of Stomatology; Changhai Hospital; Second Military Medical University; Shanghai China
| | - Xingxing Wang
- Department of Stomatology; Changhai Hospital; Second Military Medical University; Shanghai China
| | - Dalin Wang
- Department of Stomatology; Changhai Hospital; Second Military Medical University; Shanghai China
| | - Yaxian Shen
- Department of Stomatology; Changhai Hospital; Second Military Medical University; Shanghai China
| | - Da Jing
- Department of Biomedical Engineering; Fourth Military Medical University; Xi'an China
| | - Erping Luo
- Department of Biomedical Engineering; Fourth Military Medical University; Xi'an China
| | - Weizhong Tang
- Department of Stomatology; Changhai Hospital; Second Military Medical University; Shanghai China
| |
Collapse
|
44
|
Jing D, Zhai M, Tong S, Xu F, Cai J, Shen G, Wu Y, Li X, Xie K, Liu J, Xu Q, Luo E. Pulsed electromagnetic fields promote osteogenesis and osseointegration of porous titanium implants in bone defect repair through a Wnt/β-catenin signaling-associated mechanism. Sci Rep 2016; 6:32045. [PMID: 27555216 PMCID: PMC4995433 DOI: 10.1038/srep32045] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Accepted: 08/02/2016] [Indexed: 11/09/2022] Open
Abstract
Treatment of osseous defects remains a formidable clinical challenge. Porous titanium alloys (pTi) have been emerging as ideal endosseous implants due to the excellent biocompatibility and structural properties, whereas inadequate osseointegration poses risks for unreliable long-term implant stability. Substantial evidence indicates that pulsed electromagnetic fields (PEMF), as a safe noninvasive method, inhibit osteopenia/osteoporosis experimentally and clinically. We herein investigated the efficiency and potential mechanisms of PEMF on osteogenesis and osseointegration of pTi in vitro and in vivo. We demonstrate that PEMF enhanced cellular attachment and proliferation, and induced well-organized cytoskeleton for in vitro osteoblasts seeded in pTi. PEMF promoted gene expressions in Runx2, OSX, COL-1 and Wnt/β-catenin signaling. PEMF-stimulated group exhibited higher Runx2, Wnt1, Lrp6 and β-catenin protein expressions. In vivo results via μCT and histomorphometry show that 6-week and 12-week PEMF promoted osteogenesis, bone ingrowth and bone formation rate of pTi in rabbit femoral bone defect. PEMF promoted femoral gene expressions of Runx2, BMP2, OCN and Wnt/β-catenin signaling. Together, we demonstrate that PEMF improve osteogenesis and osseointegration of pTi by promoting skeletal anabolic activities through a Wnt/β-catenin signaling-associated mechanism. PEMF might become a promising biophysical modality for enhancing the repair efficiency and quality of pTi in bone defect.
Collapse
Affiliation(s)
- Da Jing
- Department of Biomedical Engineering, Fourth Military Medical University, Xi'an, China
| | - Mingming Zhai
- Department of Biomedical Engineering, Fourth Military Medical University, Xi'an, China
| | - Shichao Tong
- Department of Biomedical Engineering, Fourth Military Medical University, Xi'an, China
| | - Fei Xu
- Department of Radiation Oncology, PLA 302 Hospital, Beijing, China
| | - Jing Cai
- Department of Endocrinology, Xijing hospital, Fourth Military Medical University, Xi'an, China
| | - Guanghao Shen
- Department of Biomedical Engineering, Fourth Military Medical University, Xi'an, China
| | - Yan Wu
- Institute of Orthopaedics, Xijing hospital, Fourth Military Medical University, Xi'an, China
| | - Xiaokang Li
- Institute of Orthopaedics, Xijing hospital, Fourth Military Medical University, Xi'an, China
| | - Kangning Xie
- Department of Biomedical Engineering, Fourth Military Medical University, Xi'an, China
| | - Juan Liu
- Department of Biomedical Engineering, Fourth Military Medical University, Xi'an, China
| | - Qiaoling Xu
- Department of Nursing, Fourth Military Medical University, Xi'an, China
| | - Erping Luo
- Department of Biomedical Engineering, Fourth Military Medical University, Xi'an, China
| |
Collapse
|
45
|
Cosoli G, Scalise L, Tricarico G, Tomasini EP, Cerri G. An innovative therapy for peri-implantitis based on radio frequency electric current: numerical simulation results and clinical evidence. ANNUAL INTERNATIONAL CONFERENCE OF THE IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. ANNUAL INTERNATIONAL CONFERENCE 2016; 2016:5652-5655. [PMID: 28269537 DOI: 10.1109/embc.2016.7592009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Peri-implantitis is a severe inflammatory pathology that affects soit and hard tissues surrounding dental implants. Nowadays, only prevention is effective to contrast peri-implantitis, but, in recent years, there is the clinical evidence of the efficiency of a therapy based on the application of radio frequency electric current, reporting that 81% of the cases (66 implants, 46 patients) were successfully treated. The aim of this paper is to present the therapy mechanism, exploring the distribution of the electric currents in normal and pathologic tissues. A 3D numerical FEM model of tooth root with a dental implant screwed in the alveolar bone has been realized and the therapy has been simulated in COMSOL Multiphysics® environment. Results show that the electric current is focused in the inflamed zone around the implant, due to the fact that its conductivity is higher than the healthy tissue one. Moreover, by means of a movable return electrode, the electric current and field lines can be guided in the most inflamed area, limiting the interference on healthy tissues and improving the therapy in the area of interest. In conclusion, it can be stated that this innovative therapy would make a personalized therapy for peri-implantitis possible, also through impedance measurements, allowing the clinician to evaluate the tissue inflammation state.
Collapse
|
46
|
Kumar S, Dey S, Jain S. Extremely low-frequency electromagnetic fields: A possible non-invasive therapeutic tool for spinal cord injury rehabilitation. Electromagn Biol Med 2016; 36:88-101. [PMID: 27399648 DOI: 10.1080/15368378.2016.1194290] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Traumatic insults to the spinal cord induce both immediate mechanical damage and subsequent tissue degeneration. The latter involves a range of events namely cellular disturbance, homeostatic imbalance, ionic and neurotransmitters derangement that ultimately result in loss of sensorimotor functions. The targets for improving function after spinal cord injury (SCI) are mainly directed toward limiting these secondary injury events. Extremely low-frequency electromagnetic field (ELF-EMF) is a possible non-invasive therapeutic intervention for SCI rehabilitation which has the potential to constrain the secondary injury-induced events. In the present review, we discuss the effects of ELF-EMF on experimental and clinical SCI as well as on biological system.
Collapse
Affiliation(s)
- Suneel Kumar
- a Department of Physiology , All India Institute of Medical Sciences , New Delhi , India.,b W. M. Keck Center for Collaborative Neuroscience, Rutgers, The State University of New Jersey , Piscataway , NJ , USA
| | - Soumil Dey
- a Department of Physiology , All India Institute of Medical Sciences , New Delhi , India
| | - Suman Jain
- a Department of Physiology , All India Institute of Medical Sciences , New Delhi , India
| |
Collapse
|
47
|
Bique AM, Kaivosoja E, Mikkonen M, Paulasto-Kröckel M. Choice of osteoblast model critical for studying the effects of electromagnetic stimulation on osteogenesis in vitro. Electromagn Biol Med 2016; 35:353-64. [PMID: 27355896 DOI: 10.3109/15368378.2016.1138124] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The clinical benefits of electromagnetic field (EMF) therapy in enhancing osteogenesis have been acknowledged for decades, but agreement regarding the underlying mechanisms continues to be sought. Studies have shown EMFs to promote osteoblast-like cell proliferation, or contrarily, to induce differentiation and enhance mineralization. Typically these disparities have been attributed to methodological differences. The present paper argues the possibility that the chosen osteoblast model impacts stimulation outcome. Phenotypically immature cells, particularly at low seeding densities, appear to be prone to EMF-amplified proliferation. Conversely, mature cells at higher densities seem to be predisposed to earlier onset differentiation and mineralization. This suggests that EMFs augment ongoing processes in cell populations. To test this hypothesis, mature SaOS-2 cells and immature MC3T3-E1 cells at various densities, with or without osteo-induction, were exposed to sinusoidal 50 Hz EMF. The exposure stimulated the proliferation of MC3T3-E1 and inhibited the proliferation of SaOS-2 cells. Baseline alkaline phosphatase (ALP) expression of SaOS-2 cells was high and rapidly further increased with EMF exposure, whereas ALP effects in MC3T3-E1 cells were not seen until the second week. Thus both cell types responded differently to EMF stimulation, corroborating the hypothesis that the phenotypic maturity and culture stage of cells influence stimulation outcome.
Collapse
Affiliation(s)
- Anna-Maria Bique
- a Aalto University Department of Electrical Engineering and Automation , School of Electrical Engineering , Espoo , Finland
| | - Emilia Kaivosoja
- a Aalto University Department of Electrical Engineering and Automation , School of Electrical Engineering , Espoo , Finland
| | - Marko Mikkonen
- a Aalto University Department of Electrical Engineering and Automation , School of Electrical Engineering , Espoo , Finland
| | - Mervi Paulasto-Kröckel
- a Aalto University Department of Electrical Engineering and Automation , School of Electrical Engineering , Espoo , Finland
| |
Collapse
|
48
|
Wang R, Wu H, Yang Y, Song M. Effects of electromagnetic fields on osteoporosis: A systematic literature review. Electromagn Biol Med 2016; 35:384-90. [PMID: 27356174 DOI: 10.3109/15368378.2015.1107840] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Electromagnetic fields (EMFs) as a safe, effective and noninvasive treatment have been researched and used for many years in orthopedics, and the common use clinically is to promote fracture healing. The effects of EMFs on osteoporosis have not been well concerned. The balance between osteoblast and osteoclast activity as well as the balance between osteogenic differentiation and adipogenic differentiation of bone marrow mesenchymal stem cells plays an important role in the process of osteoporosis. A number of recent reports suggest that EMFs have a positive impact on the balances. In this review, we discuss the recent advances of EMFs in the treatment of osteoporosis from basic research to clinical study and introduce the possible mechanism. In addition, we presented future perspectives of application of EMFs for osteoporosis.
Collapse
Affiliation(s)
- Rong Wang
- a Department of Gastroenterology , General Hospital of the Yangtse River Shipping , Hubei , P. R. China
| | - Hua Wu
- b Department of Orthopedics , Tongji Hospital, Huazhong University of Science and Technology , Hubei , P. R. China
| | - Yong Yang
- b Department of Orthopedics , Tongji Hospital, Huazhong University of Science and Technology , Hubei , P. R. China
| | - Mingyu Song
- c Department of Obstetrics and Gynecology , Tongji Hospital, Huazhong University of Science and Technology , Hubei , P. R. China
| |
Collapse
|
49
|
Maziarz A, Kocan B, Bester M, Budzik S, Cholewa M, Ochiya T, Banas A. How electromagnetic fields can influence adult stem cells: positive and negative impacts. Stem Cell Res Ther 2016; 7:54. [PMID: 27086866 PMCID: PMC4834823 DOI: 10.1186/s13287-016-0312-5] [Citation(s) in RCA: 92] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
The electromagnetic field (EMF) has a great impact on our body. It has been successfully used in physiotherapy for the treatment of bone disorders and osteoarthritis, as well as for cartilage regeneration or pain reduction. Recently, EMFs have also been applied in in vitro experiments on cell/stem cell cultures. Stem cells reside in almost all tissues within the human body, where they exhibit various potential. These cells are of great importance because they control homeostasis, regeneration, and healing. Nevertheless, stem cells when become cancer stem cells, may influence the pathological condition. In this article we review the current knowledge on the effects of EMFs on human adult stem cell biology, such as proliferation, the cell cycle, or differentiation. We present the characteristics of the EMFs used in miscellaneous assays. Most research has so far been performed during osteogenic and chondrogenic differentiation of mesenchymal stem cells. It has been demonstrated that the effects of EMF stimulation depend on the intensity and frequency of the EMF and the time of exposure to it. However, other factors may affect these processes, such as growth factors, reactive oxygen species, and so forth. Exploration of this research area may enhance the development of EMF-based technologies used in medical applications and thereby improve stem cell-based therapy and tissue engineering.
Collapse
Affiliation(s)
- Aleksandra Maziarz
- Laboratory of Stem Cells' Biology, Department of Immunology, Chair of Molecular Medicine, Faculty of Medicine, University of Rzeszow, ul. Kopisto 2a, 35-310, Rzeszow, Poland.,Centre for Innovative Research in Medical and Natural Sciences, Faculty of Medicine, University of Rzeszow, ul. Warzywna 1a, 35-310, Rzeszow, Poland
| | - Beata Kocan
- Laboratory of Stem Cells' Biology, Department of Immunology, Chair of Molecular Medicine, Faculty of Medicine, University of Rzeszow, ul. Kopisto 2a, 35-310, Rzeszow, Poland.,Centre for Innovative Research in Medical and Natural Sciences, Faculty of Medicine, University of Rzeszow, ul. Warzywna 1a, 35-310, Rzeszow, Poland
| | - Mariusz Bester
- Department of Biophysics, Faculty of Mathematics and Natural Sciences, University of Rzeszow, ul. Pigonia 1, 35-310, Rzeszow, Poland
| | - Sylwia Budzik
- Department of Biophysics, Faculty of Mathematics and Natural Sciences, University of Rzeszow, ul. Pigonia 1, 35-310, Rzeszow, Poland
| | - Marian Cholewa
- Department of Biophysics, Faculty of Mathematics and Natural Sciences, University of Rzeszow, ul. Pigonia 1, 35-310, Rzeszow, Poland
| | - Takahiro Ochiya
- Division of Molecular and Cellular Medicine, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, 104-0045, Tokyo, Japan
| | - Agnieszka Banas
- Laboratory of Stem Cells' Biology, Department of Immunology, Chair of Molecular Medicine, Faculty of Medicine, University of Rzeszow, ul. Kopisto 2a, 35-310, Rzeszow, Poland. .,Centre for Innovative Research in Medical and Natural Sciences, Faculty of Medicine, University of Rzeszow, ul. Warzywna 1a, 35-310, Rzeszow, Poland.
| |
Collapse
|
50
|
Zhai M, Jing D, Tong S, Wu Y, Wang P, Zeng Z, Shen G, Wang X, Xu Q, Luo E. Pulsed electromagnetic fields promote in vitro osteoblastogenesis through a Wnt/β-catenin signaling-associated mechanism. Bioelectromagnetics 2016; 37:152-162. [PMID: 26891468 DOI: 10.1002/bem.21961] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Accepted: 01/30/2016] [Indexed: 12/18/2022]
Abstract
Substantial evidence indicates that pulsed electromagnetic fields (PEMF) could accelerate fracture healing and enhance bone mass, whereas the unclear mechanism by which PEMF stimulation promotes osteogenesis limits its extensive clinical application. In the present study, effects and potential molecular signaling mechanisms of PEMF on in vitro osteoblasts were systematically investigated. Osteoblast-like MC3T3-E1 cells were exposed to PEMF burst (0.5, 1, 2, or 6 h/day) with 15.38 Hz at various intensities (5 Gs (0.5 mT), 10 Gs (1 mT), or 20 Gs (2 mT)) for 3 consecutive days. PEMF stimulation at 20 Gs (2 mT) for 2 h/day exhibited most prominent promotive effects on osteoblastic proliferation via Cell Counting kit-8 analyses. PEMF exposure induced well-organized cytoskeleton, and promoted formation of extracellular matrix mineralization nodules. Significantly increased proliferation-related gene expressions at the proliferation phase were observed after PEMF stimulation, including Ccnd 1 and Ccne 1. PEMF resulted in significantly increased gene and protein expressions of alkaline phosphatase and osteocalcin at the differentiation phase of osteoblasts rather than the proliferation phase via quantitative reverse transcription polymerase chain reaction and Western blotting analyses. Moreover, PEMF upregulated gene and protein expressions of collagen type 1, Runt-related transcription factor 2 and Wnt/β-catenin signaling (Wnt1, Lrp6, and β-catenin) at proliferation and differentiation phases. Together, our present findings highlight that PEMF stimulated osteoblastic functions through a Wnt/β-catenin signaling-associated mechanism and, hence, regulates downstream osteogenesis-associated gene/protein expressions. Bioelectromagnetics. 37:152-162, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Mingming Zhai
- Department of Biomedical Engineering, Fourth Military Medical University, Xi'an, China
| | - Da Jing
- Department of Biomedical Engineering, Fourth Military Medical University, Xi'an, China
| | - Shichao Tong
- Department of Biomedical Engineering, Fourth Military Medical University, Xi'an, China
| | - Yan Wu
- Institute of Orthopaedics, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Pan Wang
- Department of Biomedical Engineering, Fourth Military Medical University, Xi'an, China
| | - Zhaobin Zeng
- Department of Stomatology, General Hospital of Shenyang Military Area Command, Shenyang, China
| | - Guanghao Shen
- Department of Biomedical Engineering, Fourth Military Medical University, Xi'an, China
| | - Xin Wang
- Department of Preventive Medicine, Fourth Military Medical University, Xi'an, China
| | - Qiaoling Xu
- Department of Nursing, Fourth Military Medical University, Xi'an, China
| | - Erping Luo
- Department of Biomedical Engineering, Fourth Military Medical University, Xi'an, China
| |
Collapse
|