1
|
Li C, Zhu A, Yang L, Wang X, Guo Z. Advances in magnetoelectric composites for promoting bone regeneration: a review. J Mater Chem B 2024; 12:4361-4374. [PMID: 38639047 DOI: 10.1039/d3tb02617e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/20/2024]
Abstract
Repair of large bone defects is one of the clinical problems that have not yet been fully solved. The dynamic balance of bone tissue is regulated by many biological, chemical and physical environmental factors. Simulating the microenvironment of bone tissue in the physiological state through biomimetic materials is an important development direction of tissue engineering in recent years. With the deepening of research, it has been found that when bone tissue is damaged, its surrounding magnetoelectric microenvironment is subsequently destroyed, and providing a magnetoelectric microenvironment in the biomimetic state will be beneficial to promote bone repair. This review describes the piezoelectric effect of natural bone tissue with magnetoelectric stimulation for bone regeneration, provides a detailed account of the historical development of magnetoelectric composites and the current magnetoelectric composites that are most commonly utilized in the field of tissue engineering. Besides, the hypothesized mechanistic pathways through which magnetoelectric composite materials promote bone regeneration are critically examined, including the enhancement of osteogenesis, promotion of cell adhesion and angiogenesis, modulation of bone immunity, and promotion of nerve regeneration.
Collapse
Affiliation(s)
- Chengyu Li
- Department of Periodontology and Implantology, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, P. R. China.
| | - Andi Zhu
- Department of Implantology and Prosthodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, P. R. China
| | - Liqing Yang
- Department of Periodontology and Implantology, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, P. R. China.
| | - Xinyi Wang
- Department of Periodontology and Implantology, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, P. R. China.
| | - Zehong Guo
- Department of Periodontology and Implantology, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, P. R. China.
| |
Collapse
|
2
|
Daou F, Masante B, Gabetti S, Mochi F, Putame G, Zenobi E, Scatena E, Dell'Atti F, Favero F, Leigheb M, Del Gaudio C, Bignardi C, Massai D, Cochis A, Rimondini L. Unraveling the transcriptome profile of pulsed electromagnetic field stimulation in bone regeneration using a bioreactor-based investigation platform. Bone 2024; 182:117065. [PMID: 38428556 DOI: 10.1016/j.bone.2024.117065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 02/07/2024] [Accepted: 02/26/2024] [Indexed: 03/03/2024]
Abstract
INTRODUCTION Human mesenchymal stem cells (hMSCs) sense and respond to biomechanical and biophysical stimuli, yet the involved signaling pathways are not fully identified. The clinical application of biophysical stimulation including pulsed electromagnetic field (PEMF) has gained momentum in musculoskeletal disorders and bone tissue engineering. METHODOLOGY We herein aim to explore the role of PEMF stimulation in bone regeneration by developing trabecular bone-like tissues, and then, culturing them under bone-like mechanical stimulation in an automated perfusion bioreactor combined with a custom-made PEMF stimulator. After selecting the optimal cell seeding and culture conditions for inspecting the effects of PEMF on hMSCs, transcriptomic studies were performed on cells cultured under direct perfusion with and without PEMF stimulation. RESULTS We were able to identify a set of signaling pathways and upstream regulators associated with PEMF stimulation and to distinguish those linked to bone regeneration. Our findings suggest that PEMF induces the immune potential of hMSCs by activating and inhibiting various immune-related pathways, such as macrophage classical activation and MSP-RON signaling in macrophages, respectively, while promoting angiogenesis and osteogenesis, which mimics the dynamic interplay of biological processes during bone healing. CONCLUSIONS Overall, the adopted bioreactor-based investigation platform can be used to investigate the impact of PEMF stimulation on bone regeneration.
Collapse
Affiliation(s)
- Farah Daou
- Dept. of Health Sciences, Center for Translational Research on Autoimmune and Allergic Diseases (CAAD), Università del Piemonte Orientale (UPO), Novara, Italy
| | - Beatrice Masante
- Dept. of Mechanical and Aerospace Engineering, PolitoBIOMed Lab, Politecnico di Torino, Turin, Italy; Interuniversity Center for the Promotion of the 3Rs Principles in Teaching and Research, Pisa, Italy
| | - Stefano Gabetti
- Dept. of Mechanical and Aerospace Engineering, PolitoBIOMed Lab, Politecnico di Torino, Turin, Italy; Interuniversity Center for the Promotion of the 3Rs Principles in Teaching and Research, Pisa, Italy
| | | | - Giovanni Putame
- Dept. of Mechanical and Aerospace Engineering, PolitoBIOMed Lab, Politecnico di Torino, Turin, Italy; Interuniversity Center for the Promotion of the 3Rs Principles in Teaching and Research, Pisa, Italy
| | - Eleonora Zenobi
- Hypatia Research Consortium, Rome, Italy; E. Amaldi Foundation, Rome, Italy
| | - Elisa Scatena
- Hypatia Research Consortium, Rome, Italy; E. Amaldi Foundation, Rome, Italy
| | - Federica Dell'Atti
- Dept. of Health Sciences, Center for Translational Research on Autoimmune and Allergic Diseases (CAAD), Università del Piemonte Orientale (UPO), Novara, Italy
| | - Francesco Favero
- Dept. of Health Sciences, Center for Translational Research on Autoimmune and Allergic Diseases (CAAD), Università del Piemonte Orientale (UPO), Novara, Italy
| | - Massimiliano Leigheb
- Dept. of Health Sciences, Center for Translational Research on Autoimmune and Allergic Diseases (CAAD), Università del Piemonte Orientale (UPO), Novara, Italy; Department of Orthopaedics and Traumatology, "Maggiore della Carità" Hospital, Novara, Italy
| | | | - Cristina Bignardi
- Dept. of Mechanical and Aerospace Engineering, PolitoBIOMed Lab, Politecnico di Torino, Turin, Italy; Interuniversity Center for the Promotion of the 3Rs Principles in Teaching and Research, Pisa, Italy
| | - Diana Massai
- Dept. of Mechanical and Aerospace Engineering, PolitoBIOMed Lab, Politecnico di Torino, Turin, Italy; Interuniversity Center for the Promotion of the 3Rs Principles in Teaching and Research, Pisa, Italy
| | - Andrea Cochis
- Dept. of Health Sciences, Center for Translational Research on Autoimmune and Allergic Diseases (CAAD), Università del Piemonte Orientale (UPO), Novara, Italy
| | - Lia Rimondini
- Dept. of Health Sciences, Center for Translational Research on Autoimmune and Allergic Diseases (CAAD), Università del Piemonte Orientale (UPO), Novara, Italy.
| |
Collapse
|
3
|
Du T, Liu J, Dong J, Xie H, Wang X, Yang X, Yang Y. Multifunctional coatings of nickel-titanium implant toward promote osseointegration after operation of bone tumor and clinical application: a review. Front Bioeng Biotechnol 2024; 12:1325707. [PMID: 38444648 PMCID: PMC10912669 DOI: 10.3389/fbioe.2024.1325707] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2023] [Accepted: 01/29/2024] [Indexed: 03/07/2024] Open
Abstract
Metal implants, especially Ni-Ti shape memory alloy (Ni-Ti SMA) implants, have increasingly become the first choice for fracture and massive bone defects after orthopedic bone tumor surgery. In this paper, the internal composition and shape memory properties of Ni-Ti shape memory alloy were studied. In addition, the effects of porous Ni-Ti SMA on osseointegration, and the effects of surface hydrophobicity and hydrophilicity on the osseointegration of Ni-Ti implants were also investigated. In addition, the effect of surface coating modification technology of Ni-Ti shape memory alloy on bone bonding was also studied. Several kinds of Ni-Ti alloy implants commonly used in orthopedic clinic and their advantages and disadvantages were introduced. The surface changes of Ni-Ti alloy implants promote bone fusion, enhance the adhesion of red blood cells and platelets, promote local tissue regeneration and fracture healing. In the field of orthopaedics, the use of Ni-Ti shape memory alloy implants significantly promoted clinical development. Due to the introduction of the coating, the osseointegration and biocompatibility of the implant surface have been enhanced, and the success rate of the implant has been greatly improved.
Collapse
Affiliation(s)
- Tianhao Du
- Department of Rehabilitation Medicine, General Hospital of Northern Theater Command, Shenyang, China
- Liaoning University of traditional Chinese Medicine, Shenyang, China
| | - Jia Liu
- Liaoning University of traditional Chinese Medicine, Shenyang, China
| | - Jinhan Dong
- Liaoning University of traditional Chinese Medicine, Shenyang, China
| | - Haoxu Xie
- Department of Rehabilitation Medicine, General Hospital of Northern Theater Command, Shenyang, China
- Liaoning University of traditional Chinese Medicine, Shenyang, China
| | - Xiao Wang
- Department of Rehabilitation Medicine, General Hospital of Northern Theater Command, Shenyang, China
- Liaoning University of traditional Chinese Medicine, Shenyang, China
| | - Xu Yang
- Liaoning University of traditional Chinese Medicine, Shenyang, China
| | - Yingxin Yang
- Liaoning University of traditional Chinese Medicine, Shenyang, China
| |
Collapse
|
4
|
Maiullari S, Cicirelli A, Picerno A, Giannuzzi F, Gesualdo L, Notarnicola A, Sallustio F, Moretti B. Pulsed Electromagnetic Fields Induce Skeletal Muscle Cell Repair by Sustaining the Expression of Proteins Involved in the Response to Cellular Damage and Oxidative Stress. Int J Mol Sci 2023; 24:16631. [PMID: 38068954 PMCID: PMC10706358 DOI: 10.3390/ijms242316631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2023] [Revised: 11/09/2023] [Accepted: 11/17/2023] [Indexed: 12/18/2023] Open
Abstract
Pulsed electromagnetic fields (PEMF) are employed as a non-invasive medicinal therapy, especially in the orthopedic field to stimulate bone regeneration. However, the effect of PEMF on skeletal muscle cells (SkMC) has been understudied. Here, we studied the potentiality of 1.5 mT PEMF to stimulate early regeneration of human SkMC. We showed that human SkMC stimulated with 1.5 mT PEMF for four hours repeated for two days can stimulate cell proliferation without inducing cell apoptosis or significant impairment of the metabolic activity. Interestingly, when we simulated physical damage of the muscle tissue by a scratch, we found that the same PEMF treatment can speed up the regenerative process, inducing a more complete cell migration to close the scratch and wound healing. Moreover, we investigated the molecular pattern induced by PEMF among 26 stress-related cell proteins. We found that the expression of 10 proteins increased after two consecutive days of PEMF stimulation for 4 h, and most of them were involved in response processes to oxidative stress. Among these proteins, we found that heat shock protein 70 (HSP70), which can promote muscle recovery, inhibits apoptosis and decreases inflammation in skeletal muscle, together with thioredoxin, paraoxonase, and superoxide dismutase (SOD2), which can also promote skeletal muscle regeneration following injury. Altogether, these data support the possibility of using PEMF to increase SkMC regeneration and, for the first time, suggest a possible molecular mechanism, which consists of sustaining the expression of antioxidant enzymes to control the important inflammatory and oxidative process occurring following muscle damage.
Collapse
Affiliation(s)
- Silvia Maiullari
- Department of Interdisciplinary Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy; (S.M.); (A.C.); (A.P.); (F.G.)
| | - Antonella Cicirelli
- Department of Interdisciplinary Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy; (S.M.); (A.C.); (A.P.); (F.G.)
| | - Angela Picerno
- Department of Interdisciplinary Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy; (S.M.); (A.C.); (A.P.); (F.G.)
| | - Francesca Giannuzzi
- Department of Interdisciplinary Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy; (S.M.); (A.C.); (A.P.); (F.G.)
| | - Loreto Gesualdo
- Nephrology, Dialysis and Transplantation Unit, Department of Precision and Regenerative Medicine and Ionian Area (DIMEPRE-J), University of Bari, Piazza G. Cesare 11, 70124 Bari, Italy;
| | - Angela Notarnicola
- Orthopaedic and Trauma Unit, Department of Translational Biomedicine and Neuroscience “DiBraiN”, University of Bari “Aldo Moro”, Piazza G. Cesare 11, 70124 Bari, Italy; (A.N.); (B.M.)
| | - Fabio Sallustio
- Nephrology, Dialysis and Transplantation Unit, Department of Precision and Regenerative Medicine and Ionian Area (DIMEPRE-J), University of Bari, Piazza G. Cesare 11, 70124 Bari, Italy;
| | - Biagio Moretti
- Orthopaedic and Trauma Unit, Department of Translational Biomedicine and Neuroscience “DiBraiN”, University of Bari “Aldo Moro”, Piazza G. Cesare 11, 70124 Bari, Italy; (A.N.); (B.M.)
| |
Collapse
|
5
|
Di Martino A, Villari E, Poluzzi R, Brunello M, Rossomando V, D’Agostino C, Ruta F, Faldini C. Role of biophysical stimulation in multimodal management of vertebral compression fractures. Comput Struct Biotechnol J 2023; 21:5650-5661. [PMID: 38047233 PMCID: PMC10692617 DOI: 10.1016/j.csbj.2023.11.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 11/10/2023] [Accepted: 11/10/2023] [Indexed: 12/05/2023] Open
Abstract
Raised life expectancy and aging of the general population are associated with an increased concern for fragility fractures due to factors such as osteoporosis, reduced bone density, and an higher risk of falls. Among these, the most frequent are vertebral compression fractures (VCF), which can be clinically occult. Once the diagnosis is made, generally thorough antero-posterior and lateral views of the affected spine at the radiographs, a comprehensive workup to assess the presence of a metabolic bone disease or secondary causes of osteoporosis and bone frailty is required. Treatment uses a multimodal management consisting of a combination of brace, pain management, bone metabolism evaluation, osteoporosis medication and has recently incorporated biophysical stimulation, a noninvasive technique that uses induced electric stimulation to improve bone recovery through the direct and indirect upregulation of bone morphogenic proteins, stimulating bone formation and remodeling. It contributes to the effectiveness of the therapy, promoting accelerated healing, supporting the reduction of bed rest and pain medications, improving patients' quality of life, and reducing the risk to undergo surgery in patients affected by VCFs. Therefore, the aim of this review is to outline the fundamental concepts of multimodal treatment for VCF, as well as the present function and significance of biophysical stimulation in the treatment of VCF patients.
Collapse
Affiliation(s)
- Alberto Di Martino
- 1st Orthopaedic and Traumatologic Department, IRCCS Istituto Ortopedico Rizzoli, Via G.B. Pupilli 1, 40136 Bologna, Italy
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Italy
| | - Eleonora Villari
- 1st Orthopaedic and Traumatologic Department, IRCCS Istituto Ortopedico Rizzoli, Via G.B. Pupilli 1, 40136 Bologna, Italy
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Italy
| | - Riccardo Poluzzi
- 1st Orthopaedic and Traumatologic Department, IRCCS Istituto Ortopedico Rizzoli, Via G.B. Pupilli 1, 40136 Bologna, Italy
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Italy
| | - Matteo Brunello
- 1st Orthopaedic and Traumatologic Department, IRCCS Istituto Ortopedico Rizzoli, Via G.B. Pupilli 1, 40136 Bologna, Italy
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Italy
| | - Valentino Rossomando
- 1st Orthopaedic and Traumatologic Department, IRCCS Istituto Ortopedico Rizzoli, Via G.B. Pupilli 1, 40136 Bologna, Italy
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Italy
| | - Claudio D’Agostino
- 1st Orthopaedic and Traumatologic Department, IRCCS Istituto Ortopedico Rizzoli, Via G.B. Pupilli 1, 40136 Bologna, Italy
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Italy
| | - Federico Ruta
- 1st Orthopaedic and Traumatologic Department, IRCCS Istituto Ortopedico Rizzoli, Via G.B. Pupilli 1, 40136 Bologna, Italy
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Italy
| | - Cesare Faldini
- 1st Orthopaedic and Traumatologic Department, IRCCS Istituto Ortopedico Rizzoli, Via G.B. Pupilli 1, 40136 Bologna, Italy
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Italy
| |
Collapse
|
6
|
Hao X, Wang D, Yan Z, Ding Y, Zhang J, Liu J, Shao X, Liu X, Wang L, Luo E, Cai J, Jing D. Bone Deterioration in Response to Chronic High-Altitude Hypoxia Is Attenuated by a Pulsed Electromagnetic Field Via the Primary Cilium/HIF-1α Axis. J Bone Miner Res 2023; 38:597-614. [PMID: 36680558 DOI: 10.1002/jbmr.4772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 12/14/2022] [Accepted: 01/07/2023] [Indexed: 01/22/2023]
Abstract
Chronic high-altitude hypoxia induces irreversible abnormalities in various organisms. Emerging evidence indicates that hypobaric hypoxia markedly suppresses bone mass and bone strength. However, few effective means have been identified to prevent such bone deficits. Here, we assessed the potential of pulsed electromagnetic fields (PEMFs) to noninvasively resist bone deterioration induced by hypobaric hypoxia. We observed that exogenous PEMF treatment at 15 Hz and 20 Gauss (Gs) improved the cancellous and cortical bone mass, bone microstructure, and skeletal mechano-properties in rats subjected to chronic exposure of hypobaric hypoxia simulating an altitude of 4500 m for 6 weeks by primarily modulating osteoblasts and osteoblast-mediated bone-forming activity. Moreover, our results showed that whereas PEMF stimulated the functional activity of primary osteoblasts in hypoxic culture in vitro, it had negligible effects on osteoclasts and osteocytes exposed to hypoxia. Mechanistically, the primary cilium was found to function as the major electromagnetic sensor in osteoblasts exposed to hypoxia. The polycystins PC-1/PC-2 complex was identified as the primary calcium channel in the primary cilium of hypoxia-exposed osteoblastic cells responsible for the detection of external PEMF signals, and thereby translated these biophysical signals into intracellular biochemical events involving significant increase in the intracellular soluble adenylyl cyclase (sAC) expression and subsequent elevation of cyclic adenosine monophosphate (cAMP) concentration. The second messenger cAMP inhibited the transcription of oxygen homeostasis-related hypoxia-inducible factor 1-alpha (HIF-1α), and thus enhanced osteoblast differentiation and improved bone phenotype. Overall, the present study not only advances our understanding of bone physiology at high altitudes, but more importantly, proposes effective means to ameliorate high altitude-induced bone loss in a noninvasive and cost-effective manner. © 2023 American Society for Bone and Mineral Research (ASBMR).
Collapse
Affiliation(s)
- Xiaoxia Hao
- School of Life Science, Northwest University, Xi'an, China.,Department of Biomedical Engineering, Fourth Military Medical University, Xi'an, China
| | - Dan Wang
- School of Life Science, Northwest University, Xi'an, China.,Department of Biomedical Engineering, Fourth Military Medical University, Xi'an, China
| | - Zedong Yan
- Department of Biomedical Engineering, Fourth Military Medical University, Xi'an, China
| | - Yuanjun Ding
- Department of Biomedical Engineering, Fourth Military Medical University, Xi'an, China
| | - Juan Zhang
- School of Life Science, Northwest University, Xi'an, China
| | - Juan Liu
- Department of Biomedical Engineering, Fourth Military Medical University, Xi'an, China
| | - Xi Shao
- Department of Biomedical Engineering, Fourth Military Medical University, Xi'an, China
| | - Xiyu Liu
- Department of Biomedical Engineering, Fourth Military Medical University, Xi'an, China
| | - Lu Wang
- School of Life Science, Northwest University, Xi'an, China
| | - Erping Luo
- Department of Biomedical Engineering, Fourth Military Medical University, Xi'an, China
| | - Jing Cai
- College of Basic Medicine, Shaanxi University of Chinese Medicine, Xianyang, China
| | - Da Jing
- Department of Biomedical Engineering, Fourth Military Medical University, Xi'an, China
| |
Collapse
|
7
|
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: 1.0] [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
|
8
|
Guo S, Zhao G, Chen XM, Xue Y, OuYang XL, Liu JY, Huang YP, Liu YJ, Yao Q, Han L, Zhang CH, Li B, Wang Q, Zhao B. Effect of transcutaneous electrical acupoint stimulation on bone metabolism in patients with immobilisation after foot and ankle fracture surgery: a randomised controlled trial study protocol. BMJ Open 2022; 12:e056691. [PMID: 36691208 PMCID: PMC9462119 DOI: 10.1136/bmjopen-2021-056691] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Accepted: 08/08/2022] [Indexed: 01/26/2023] Open
Abstract
INTRODUCTION Fracture is a disease with a high incidence worldwide. Foot and ankle fractures are common among fractures of the lower extremities. Foot and ankle fractures usually require surgical fixation and a period of fixed treatment, which can lead to decreased bone density. Although transcutaneous electrical acupoint stimulation (TEAS) is widely used for movement system diseases, there is minimal evidence to show the effectiveness of TEAS on patients after surgical fixation of ankle and foot fractures. This trial aims to evaluate whether TEAS can reduce bone loss in patients with immobilisation after ankle and foot fractures. METHODS AND ANALYSIS A randomised controlled trial will be conducted in which 60 patients will be randomly divided into two groups: (a) the control group will be treated according to the routine procedures of basic orthopaedics treatment; (b) in the treatment group, bilateral SP36, BL23 and ST36 will be performed on the basis of the control group, and the test will be performed for 30 min every other day for a total of 8 weeks. Bone turnover markers will be used as primary outcome. Secondary outcomes are composed of blood phosphorus, blood calcium and bone mineral density. Treatment safety will be monitored and recorded. ETHICS AND DISSEMINATION This trial is approved by the Ethics Committee of Beijing University of Chinese Medicine (2020BZYLL0611) and the Ethics Committee of Beijing Luhe Hospital (2020-LHKY-055-02), and inpatients who meet the following diagnostic and inclusion criteria are eligible to participate in this study. TRIAL REGISTRATION NUMBER ChiCTR 2000039944.
Collapse
Affiliation(s)
- Shiqi Guo
- School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing, China
| | - Guozhen Zhao
- Capital Medical University, Beijing Hospital of Traditional Chinese Medicine, Beijing, China
| | - Xue-Ming Chen
- Capital Medical University, Beijing Luhe Hospital, Beijing, China
| | - Ying Xue
- School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing, China
| | - Xia-Li OuYang
- School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing, China
| | - Jin-Yi Liu
- School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing, China
| | - Yue-Ping Huang
- School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing, China
| | - Ya-Jie Liu
- School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing, China
| | - Qin Yao
- School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing, China
| | - Li Han
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Chun-Hui Zhang
- Capital Medical University, Beijing Luhe Hospital, Beijing, China
| | - Bo Li
- Capital Medical University, Beijing Hospital of Traditional Chinese Medicine, Beijing, China
| | - Qi Wang
- Capital Medical University, Beijing Luhe Hospital, Beijing, China
| | - Baixiao Zhao
- Affiliated Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
9
|
Gerdesmeyer L, Zielhardt P, Klüter T, Gollwitzer H, Gerdesmeyer L, Hausdorf J, Ringeisen M, Knobloch K, Saxena A, Krath A. Stimulation of human bone marrow mesenchymal stem cells by electromagnetic transduction therapy - EMTT. Electromagn Biol Med 2022; 41:304-314. [DOI: 10.1080/15368378.2022.2079672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Affiliation(s)
- Ludger Gerdesmeyer
- Department of Sports Medicine, Palo Alto Medical Center, Palo Alto, USA
- Department of Orthopedic and Traumatology, Technical University Munich, Munich Germany
- Städtisches Krankenhaus Kiel, Kiel
| | - Paula Zielhardt
- Department of Orthopaedic Surgery and Traumatology, University Schleswig Holstein, Kiel, Germany
| | - Tim Klüter
- Department of Orthopaedic Surgery and Traumatology, University Schleswig Holstein, Kiel, Germany
| | - Hans Gollwitzer
- Department of Orthopedic and Traumatology, Technical University Munich, Munich Germany
| | - Lennart Gerdesmeyer
- Department of Orthopedic and Traumatology, Technical University Munich, Munich Germany
| | - Joerg Hausdorf
- Orthopedic Department, Physical Medicine and Rehabilitation, University Hospital of Munich, Germany
| | - Martin Ringeisen
- Department of Orthopedic Surgery and Traumatology, Orthopaedic Medical Center Dr. Ringeisen, Augsburg, Germany
| | - Karsten Knobloch
- Orthopedic Department, SportPraxis Prof. Dr. med. Karsten Knobloch, Hannover
| | - Amol Saxena
- Department of Sports Medicine, Palo Alto Medical Center, Palo Alto, USA
| | - André Krath
- Department of Orthopaedic Surgery and Traumatology, University Schleswig Holstein, Kiel, Germany
| |
Collapse
|
10
|
Diamagnetic Therapy in a Patient with Complex Regional Pain Syndrome Type I and Multiple Drug Intolerance: A Case Report. REPORTS 2022. [DOI: 10.3390/reports5020018] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Complex regional pain syndrome (CRPS) is a neurologic chronic pain condition hard to diagnose and treat, and able to significantly impact the quality of life. Currently, the available multimodal, individualized treatments (i.e., pharmacological and non-pharmacological therapies including invasive procedures) are aimed only at symptom control. Herein, we report a 69-year-old Caucasian female who came to our attention due to a 3-year history of severe (10/10) burning pain in her right ankle, along with oedema and local changes in skin color and temperature, which occurred after the ankle sprain. Previous pharmacological attempts failed due to multiple drug intolerance. Clinical examination confirmed the CRPS type I diagnosis, and a weekly diamagnetic therapy protocol was started since the patient refused further medications and interventional procedures. After 10 weeks of treatment, a significant (p < 0.01) reduction in pain severity and absence of oedema (difference in ankles’ circumference: from 3 cm to 0) were observed, with consequent improvements in quality of life and no adverse events. Although high-quality clinical evidence is still lacking, our case report suggests further investigating the potential use of diamagnetic therapy as a non-invasive and safe adjunctive treatment for CRPS, and as an alternative when patients did not benefit from drugs and/or refuse invasive procedures.
Collapse
|
11
|
Hamid HA, Ramasamy R, Mustafa MK, Hosseinpour Sarmadi V, Miskon A. Magnetic exposure using Samarium Cobalt (SmC O5) increased proliferation and stemness of human Umbilical Cord Mesenchymal Stem Cells (hUC-MSCs). Sci Rep 2022; 12:8904. [PMID: 35618759 PMCID: PMC9135697 DOI: 10.1038/s41598-022-12653-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 05/11/2022] [Indexed: 12/12/2022] Open
Abstract
Despite the extensive reports on the potential hazard of magnetic field (MF) exposures on humans, there are also concurrently reported on the improved proliferative property of stem cells at optimum exposure. However, the effect on mesenchymal stem cells (MSCs) remains unknown. Therefore, we aimed to investigate the impact of induced static MF (SMF) on human umbilical cord-derived mesenchymal stem cells (hUC-MSCs) using Samarium Cobalt (SmCO5). At passage 3, hUC-MSCs (1 × 104) were exposed to 21.6 mT SMF by a direct exposure (DE) showed a significantly higher cell count (p < 0.05) in the growth kinetics assays with the shortest population doubling time relative to indirect exposure and negative control. The DE group was committed into the cell cycle with increased S phase (55.18 ± 1.38%) and G2/M phase (21.75 ± 1.38%) relative to the NC group [S-phase (13.54 ± 2.73%); G2/M phase (8.36 ± 0.28%)]. Although no significant changes were observed in the immunophenotype, the DE group showed an elevated expression of pluripotency-associated markers (OCT4, SOX2, NANOG, and REX1). These results suggest that the MFs could potentially induce proliferation of MSCs, a promising approach to promote stem cells propagation for clinical therapy and research without compromising the stemness of hUC-MSCs.
Collapse
Affiliation(s)
- Haslinda Abdul Hamid
- Bio Artificial Organ and Regenerative Medicine Unit, National Defense University of Malaysia, Sungai Besi Camp, 57000, Kuala Lumpur, Malaysia
| | - Rajesh Ramasamy
- Stem Cell & Immunity Research Group, Immunology Laboratory, Department of Pathology, Faculty of Medicine and Health Sciences, University Putra Malaysia, 43400, Serdang, Malaysia.,Department of Dental Radiology, Faculty of Dental Medicine, Airlangga University, Surabaya, 60132, Indonesia
| | - Mohd Kamarulzaki Mustafa
- Department of Physics, Faculty of Applied Sciences and Technology, University Tun Hussein Onn Malaysia, Pagoh Campus, KM1, Jalan Panchor, Hub Pendidikan Tinggi Pagoh, 84600, Muar, Johor, Malaysia
| | - Vahid Hosseinpour Sarmadi
- Institutes of Regenerative Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran.,Cellular and Molecular Research Centre, Iran University of Medical Sciences, Tehran, Iran
| | - Azizi Miskon
- Bio Artificial Organ and Regenerative Medicine Unit, National Defense University of Malaysia, Sungai Besi Camp, 57000, Kuala Lumpur, Malaysia.
| |
Collapse
|
12
|
Hung CT, Racine-Avila J, Pellicore MJ, Aaron R. Biophysical Modulation of Mesenchymal Stem Cell Differentiation in the Context of Skeletal Repair. Int J Mol Sci 2022; 23:ijms23073919. [PMID: 35409277 PMCID: PMC8998876 DOI: 10.3390/ijms23073919] [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: 02/28/2022] [Revised: 03/25/2022] [Accepted: 03/29/2022] [Indexed: 11/16/2022] Open
Abstract
A prominent feature of the skeleton is its ability to remodel in response to biophysical stimuli and to repair under varied biophysical conditions. This allows the skeleton considerable adaptation to meet its physiological roles of stability and movement. Skeletal cells and their mesenchymal precursors exist in a native environment rich with biophysical signals, and they sense and respond to those signals to meet organismal demands of the skeleton. While mechanical strain is the most recognized of the skeletal biophysical stimuli, signaling phenomena also include fluid flow, hydrostatic pressure, shear stress, and ion-movement-related electrokinetic phenomena including, prominently, streaming potentials. Because of the complex interactions of these electromechanical signals, it is difficult to isolate the significance of each. The application of external electrical and electromagnetic fields allows an exploration of the effects of these stimuli on cell differentiation and extra-cellular matrix formation in the absence of mechanical strain. This review takes a distinctly translational approach to mechanistic and preclinical studies of differentiation and skeletal lineage commitment of mesenchymal cells under biophysical stimulation. In vitro studies facilitate the examination of isolated cellular responses while in vivo studies permit the observation of cell differentiation and extracellular matrix synthesis.
Collapse
Affiliation(s)
- Clark T. Hung
- Department of Biomedical Engineering, Columbia University, New York, NY 10032, USA; (C.T.H.); (M.J.P.)
- Department of Orthopedic Surgery, Columbia University, New York, NY 10032, USA
| | - Jennifer Racine-Avila
- Department of Orthopedics, Alpert Medical School of Brown University, Providence, RI 02905, USA;
| | - Matthew J. Pellicore
- Department of Biomedical Engineering, Columbia University, New York, NY 10032, USA; (C.T.H.); (M.J.P.)
| | - Roy Aaron
- Department of Orthopedics, Alpert Medical School of Brown University, Providence, RI 02905, USA;
- Correspondence: ; Tel.: +1-401-274-9660
| |
Collapse
|
13
|
Pulsed Electro-Magnetic Field (PEMF) Effect on Bone Healing in Animal Models: A Review of Its Efficacy Related to Different Type of Damage. BIOLOGY 2022; 11:biology11030402. [PMID: 35336776 PMCID: PMC8945722 DOI: 10.3390/biology11030402] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 02/18/2022] [Accepted: 03/03/2022] [Indexed: 11/17/2022]
Abstract
Simple Summary Pulsed electromagnetic fields (PEMFs) are a type of biophysical stimulation that has been shown to be effective in improving bone regeneration and preventing bone loss. Their use dates back to the 1970s, but a gold standard treatment protocol has not yet been defined. PEMF efficacy relies on the generation of biopotentials, which activate several molecular pathways. There is currently no clear understanding of the effects on bone healing and, in addition, there are several animal models relevant to this issue. Therefore, drawing guidelines and conclusions from the analysis of the studies is difficult. In vivo investigations on PEMF stimulation are reviewed in this paper, focusing on molecular and morphological improvements in bone. Currently, there is little knowledge about the biological mechanism of PEMF and its effect on bone healing. This is due to the variability of crucial characteristics of electro-magnetic fields, such as amplitude and exposure frequency, which may influence the type of biological response. Furthermore, a different responsiveness of cells involved in the bone healing process is documented. Heterogeneous setting parameters and different outcome measures are considered in various animal models. Therefore, achieving comparable results is difficult. Abstract Biophysical energies are a versatile tool to stimulate tissues by generating biopotentials. In particular, pulsed electromagnetic field (PEMF) stimulation has intrigued researchers since the 1970s. To date, many investigations have been carried out in vivo, but a gold standard treatment protocol has not yet been defined. The main obstacles are represented by the complex setting of PEMF characteristics, the variety of animal models (including direct and indirect bone damage) and the lack of a complete understanding of the molecular pathways involved. In the present review the main studies about PEMF stimulation in animal models with bone impairment were reviewed. PEMF signal characteristics were investigated, as well as their effect on molecular pathways and osseous morphological features. We believe that this review might be a useful starting point for a prospective study in a clinical setting. Consistent evidence from the literature suggests a potential beneficial role of PEMF in clinical practice. Nevertheless, the wide variability of selected parameters (frequency, duration, and amplitude) and the heterogeneity of applied protocols make it difficult to draw certain conclusions about PEMF effectiveness in clinical implementation to promote bone healing. Deepening the knowledge regarding the most consistent results reported in literature to date, we believe that this review may be a useful starting point to propose standardized experimental guidelines. This might provide a solid base for further controlled trials, to investigate PEMF efficacy in bone damage conditions during routine clinical practice.
Collapse
|
14
|
Hamid HA, Sarmadi VH, Prasad V, Ramasamy R, Miskon A. Electromagnetic field exposure as a plausible approach to enhance the proliferation and differentiation of mesenchymal stem cells in clinically relevant scenarios. J Zhejiang Univ Sci B 2022; 23:42-57. [PMID: 35029087 PMCID: PMC8758935 DOI: 10.1631/jzus.b2100443] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Mesenchymal stem/stromal cell (MSC)-based therapy has been regarded as one of the most revolutionary breakthroughs in the history of modern medicine owing to its myriad of immunoregulatory and regenerative properties. With the rapid progress in the fields of osteo- and musculoskeletal therapies, the demand for MSC-based treatment modalities is becoming increasingly prominent. In this endeavor, researchers around the world have devised new and innovative techniques to support the proliferation of MSCs while minimizing the loss of hallmark features of stem cells. One such example is electromagnetic field (EMF) exposure, which is an alternative approach with promising potential. In this review, we present a critical discourse on the efficiency, practicability, and limitations of some of the relevant methods, with insurmountable evidence backing the implementation of EMF as a feasible strategy for the clinically relevant expansion of MSCs.
Collapse
Affiliation(s)
- Haslinda Abdul Hamid
- Bio-artificial Organ and Regenerative Medicine Unit, National Defense University of Malaysia, Kuala Lumpur 57000, Malaysia
| | - Vahid Hosseinpour Sarmadi
- Cellular and Molecular Research Centre, Iran University of Medical Sciences, Tehran 144961 4535, Iran.,Institutes of Regenerative Medicine, Iran University of Medical Sciences, Tehran 199671 4353, Iran
| | - Vivek Prasad
- Stem Cell and Immunity Research Group, Immunology Laboratory, Department of Pathology, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia (UPM), Selangor 43400, Malaysia
| | - Rajesh Ramasamy
- Stem Cell and Immunity Research Group, Immunology Laboratory, Department of Pathology, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia (UPM), Selangor 43400, Malaysia
| | - Azizi Miskon
- Bio-artificial Organ and Regenerative Medicine Unit, National Defense University of Malaysia, Kuala Lumpur 57000, Malaysia.
| |
Collapse
|
15
|
Li Y, Yang Y, Wang M, Zhang X, Bai S, Lu X, Li Y, Waldorff EI, Zhang N, Lee WYW, Li G. High slew rate pulsed electromagnetic field enhances bone consolidation and shortens daily treatment duration in distraction osteogenesis. Bone Joint Res 2021; 10:767-779. [PMID: 34872332 PMCID: PMC8696558 DOI: 10.1302/2046-3758.1012.bjr-2021-0274.r1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Aims Distraction osteogenesis (DO) is a useful orthopaedic procedure employed to lengthen and reshape bones by stimulating bone formation through controlled slow stretching force. Despite its promising applications, difficulties are still encountered. Our previous study demonstrated that pulsed electromagnetic field (PEMF) treatment significantly enhances bone mineralization and neovascularization, suggesting its potential application. The current study compared a new, high slew rate (HSR) PEMF signal, with different treatment durations, with the standard Food and Drug Administration (FDA)-approved signal, to determine if HSR PEMF is a better alternative for bone formation augmentation. Methods The effects of a HSR PEMF signal with three daily treatment durations (0.5, one, and three hours/day) were investigated in an established rat DO model with comparison of an FDA-approved classic signal (three hrs/day). PEMF treatments were applied to the rats daily for 35 days, starting from the distraction phase until termination. Radiography, micro-CT (μCT), biomechanical tests, and histological examinations were employed to evaluate the quality of bone formation. Results All rats tolerated the treatment well and no obvious adverse effects were found. By comparison, the HSR signal (three hrs/day) treatment group achieved the best healing outcome, in that endochondral ossification and bone consolidation were enhanced. In addition, HSR signal treatment (one one hr/day) had similar effects to treatment using the classic signal (three three hrs/day), indicating that treatment duration could be significantly shortened with the HSR signal. Conclusion HSR signal may significantly enhance bone formation and shorten daily treatment duration in DO, making it a potential candidate for a new clinical protocol for patients undergoing DO treatments. Cite this article: Bone Joint Res 2021;10(12):767–779.
Collapse
Affiliation(s)
- Yucong Li
- Department of Orthopaedic and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China.,Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, China
| | - Yongkang Yang
- Department of Orthopaedic and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China.,Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, China
| | - Ming Wang
- Department of Orthopaedic and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China.,Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, China
| | - Xiaoting Zhang
- Department of Orthopaedic and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China.,Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, China
| | - Shanshan Bai
- Department of Orthopaedic and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China.,Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, China
| | - Xuan Lu
- Department of Orthopaedic and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China.,Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, China
| | - Yuan Li
- Department of Orthopaedic and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China.,Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, China
| | - Erik I Waldorff
- Research & Clinical Affairs, Orthofix Medical Inc, Lewisville, Texas, USA
| | - Nianli Zhang
- Research & Clinical Affairs, Orthofix Medical Inc, Lewisville, Texas, USA
| | - Wayne Yuk-Wai Lee
- Department of Orthopaedic and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China.,SH Ho Scoliosis Research Laboratory, Joint Scoliosis Research Center of the Chinese University of Hong Kong and Nanjing University, Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Hong Kong, China.,Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Gang Li
- Department of Orthopaedic and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China.,Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, China
| |
Collapse
|
16
|
Hao Z, Xu Z, Wang X, Wang Y, Li H, Chen T, Hu Y, Chen R, Huang K, Chen C, Li J. Biophysical Stimuli as the Fourth Pillar of Bone Tissue Engineering. Front Cell Dev Biol 2021; 9:790050. [PMID: 34858997 PMCID: PMC8630705 DOI: 10.3389/fcell.2021.790050] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Accepted: 10/26/2021] [Indexed: 01/12/2023] Open
Abstract
The repair of critical bone defects remains challenging worldwide. Three canonical pillars (biomaterial scaffolds, bioactive molecules, and stem cells) of bone tissue engineering have been widely used for bone regeneration in separate or combined strategies, but the delivery of bioactive molecules has several obvious drawbacks. Biophysical stimuli have great potential to become the fourth pillar of bone tissue engineering, which can be categorized into three groups depending on their physical properties: internal structural stimuli, external mechanical stimuli, and electromagnetic stimuli. In this review, distinctive biophysical stimuli coupled with their osteoinductive windows or parameters are initially presented to induce the osteogenesis of mesenchymal stem cells (MSCs). Then, osteoinductive mechanisms of biophysical transduction (a combination of mechanotransduction and electrocoupling) are reviewed to direct the osteogenic differentiation of MSCs. These mechanisms include biophysical sensing, transmission, and regulation. Furthermore, distinctive application strategies of biophysical stimuli are presented for bone tissue engineering, including predesigned biomaterials, tissue-engineered bone grafts, and postoperative biophysical stimuli loading strategies. Finally, ongoing challenges and future perspectives are discussed.
Collapse
Affiliation(s)
- Zhuowen Hao
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Zhenhua Xu
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Xuan Wang
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Yi Wang
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Hanke Li
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Tianhong Chen
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Yingkun Hu
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Renxin Chen
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Kegang Huang
- Wuhan Institute of Proactive Health Management Science, Wuhan, China
| | - Chao Chen
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Department of Orthopedics, Hefeng Central Hospital, Enshi, China
| | - Jingfeng Li
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
17
|
Barati M, Darvishi B, Javidi MA, Mohammadian A, Shariatpanahi SP, Eisavand MR, Madjid Ansari A. Cellular stress response to extremely low-frequency electromagnetic fields (ELF-EMF): An explanation for controversial effects of ELF-EMF on apoptosis. Cell Prolif 2021; 54:e13154. [PMID: 34741480 PMCID: PMC8666288 DOI: 10.1111/cpr.13154] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 09/21/2021] [Accepted: 10/12/2021] [Indexed: 12/12/2022] Open
Abstract
Impaired apoptosis is one of the hallmarks of cancer, and almost all of the non‐surgical approaches of eradicating tumour cells somehow promote induction of apoptosis. Indeed, numerous studies have stated that non‐ionizing non‐thermal extremely low‐frequency magnetic fields (ELF‐MF) can modulate the induction of apoptosis in exposed cells; however, much controversy exists in observations. When cells are exposed to ELF‐EMF alone, very low or no statistically significant changes in apoptosis are observed. Contrarily, exposure to ELF‐EMF in the presence of a co‐stressor, including a chemotherapeutic agent or ionizing radiation, can either potentiate or inhibit apoptotic effects of the co‐stressor. In our idea, the main point neglected in interpreting these discrepancies is “the cellular stress responses” of cells following ELF‐EMF exposure and its interplay with apoptosis. The main purpose of the current review was to outline the triangle of ELF‐EMF, the cellular stress response of cells and apoptosis and to interpret and unify discrepancies in results based on it. Therefore, initially, we will describe studies performed on identifying the effect of ELF‐EMF on induction/inhibition of apoptosis and enumerate proposed pathways through which ELF‐EMF exposure may affect apoptosis; then, we will explain cellular stress response and cues for its induction in response to ELF‐EMF exposure; and finally, we will explain why such controversies have been observed by different investigators.
Collapse
Affiliation(s)
- Mojdeh Barati
- Integrative Oncology Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran
| | - Behrad Darvishi
- Recombinant Proteins Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran
| | - Mohammad Amin Javidi
- Integrative Oncology Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran
| | - Ali Mohammadian
- Department of Medical Biotechnology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | | | - Mohammad Reza Eisavand
- Recombinant Proteins Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran
| | - Alireza Madjid Ansari
- Integrative Oncology Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran
| |
Collapse
|
18
|
Oltean-Dan D, Dogaru GB, Jianu EM, Riga S, Tomoaia-Cotisel M, Mocanu A, Barbu-Tudoran L, Tomoaia G. Biomimetic Composite Coatings for Activation of Titanium Implant Surfaces: Methodological Approach and In Vivo Enhanced Osseointegration. MICROMACHINES 2021; 12:mi12111352. [PMID: 34832764 PMCID: PMC8618198 DOI: 10.3390/mi12111352] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Revised: 10/24/2021] [Accepted: 10/25/2021] [Indexed: 12/14/2022]
Abstract
Innovative nanomaterials are required for the coatings of titanium (Ti) implants to ensure the activation of Ti surfaces for improved osseointegration, enhanced bone fracture healing and bone regeneration. This paper presents a systematic investigation of biomimetic composite (BC) coatings on Ti implant surfaces in a rat model of a diaphyseal femoral fracture. Methodological approaches of surface modification of the Ti implants via the usual joining methods (e.g., grit blasting and acid etching) and advanced physicochemical coating via a self-assembled dip-coating method were used. The biomimetic procedure used multi-substituted hydroxyapatite (ms-HAP) HAP-1.5 wt% Mg-0.2 wt% Zn-0.2 wt% Si nanoparticles (NPs), which were functionalized using collagen type 1 molecules (COL), resulting in ms-HAP/COL (core/shell) NPs that were embedded into a polylactic acid (PLA) matrix and finally covered with COL layers, obtaining the ms-HAP/COL@PLA/COL composite. To assess the osseointegration issue, first, the thickness, surface morphology and roughness of the BC coating on the Ti implants were determined using AFM and SEM. The BC-coated Ti implants and uncoated Ti implants were then used in Wistar albino rats with a diaphyseal femoral fracture, both in the absence and the presence of high-frequency pulsed electromagnetic shortwave (HF-PESW) stimulation. This study was performed using a bone marker serum concentration and histological and computer tomography (micro-CT) analysis at 2 and 8 weeks after surgical implantation. The implant osseointegration was evaluated through the bone–implant contact (BIC). The bone–implant interface was investigated using FE-SEM images and EDX spectra of the retrieved surgical implants at 8 weeks in the four animal groups. The obtained results showed significantly higher bone–implants contact and bone volume per tissue volume, as well as a greater amount of newly formed bone, in the BC-coated Ti implants than in the uncoated Ti implants. Direct bone–implant contact was also confirmed via histological examination. The results of this study confirmed that these biomimetic composite coatings on Ti implants were essential for a significant enhancement of osseointegration of BC-coated Ti implants and bone regeneration. This research provides a novel strategy for the treatment of bone fractures with possible orthopedic applications.
Collapse
Affiliation(s)
- Daniel Oltean-Dan
- Department of Orthopedics and Traumatology, Iuliu Hatieganu University of Medicine and Pharmacy, 47 General Traian Mosoiu Street, 400132 Cluj-Napoca, Romania;
| | - Gabriela-Bombonica Dogaru
- Department of Medical Rehabilitation, Iuliu Hatieganu University of Medicine and Pharmacy, 46-50 Viilor Street, 400347 Cluj-Napoca, Romania;
| | - Elena-Mihaela Jianu
- Department of Histology, Iuliu Hatieganu University of Medicine and Pharmacy, 6 Louis Pasteur Street, 400349 Cluj-Napoca, Romania;
| | - Sorin Riga
- Research Center of Physical Chemistry, Department of Chemical Engineering, Faculty of Chemistry and Chemical Engineering, Babes-Bolyai University, 11 Arany Janos Street, 400028 Cluj-Napoca, Romania; (S.R.); (A.M.)
- Academy of Romanian Scientists, 54 Splaiul Independentei, 050085 Bucharest, Romania
| | - Maria Tomoaia-Cotisel
- Research Center of Physical Chemistry, Department of Chemical Engineering, Faculty of Chemistry and Chemical Engineering, Babes-Bolyai University, 11 Arany Janos Street, 400028 Cluj-Napoca, Romania; (S.R.); (A.M.)
- Academy of Romanian Scientists, 54 Splaiul Independentei, 050085 Bucharest, Romania
- Correspondence: (M.T.-C.); (G.T.)
| | - Aurora Mocanu
- Research Center of Physical Chemistry, Department of Chemical Engineering, Faculty of Chemistry and Chemical Engineering, Babes-Bolyai University, 11 Arany Janos Street, 400028 Cluj-Napoca, Romania; (S.R.); (A.M.)
| | - Lucian Barbu-Tudoran
- Electron Microscopy Laboratory Prof. C. Craciun, Faculty of Biology and Geology, Babes-Bolyai University, 5-7 Clinicilor Street, 400006 Cluj-Napoca, Romania;
| | - Gheorghe Tomoaia
- Department of Orthopedics and Traumatology, Iuliu Hatieganu University of Medicine and Pharmacy, 47 General Traian Mosoiu Street, 400132 Cluj-Napoca, Romania;
- Academy of Romanian Scientists, 54 Splaiul Independentei, 050085 Bucharest, Romania
- Correspondence: (M.T.-C.); (G.T.)
| |
Collapse
|
19
|
Eid MM, El-Gendy AM, Abdelbasset WK, Elkholi SM, Abdel-fattah MS. The effect of magnetic therapy and moderate aerobic exercise on osteoporotic patients: A randomized clinical study. Medicine (Baltimore) 2021; 100:e27379. [PMID: 34596156 PMCID: PMC8483884 DOI: 10.1097/md.0000000000027379] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Accepted: 09/13/2021] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND Osteoporosis is a frequent musculoskeletal condition with significant complications that would be a global health problem and one of the major causes of mortality and morbidity. OBJECTIVES The current study aimed to ascertain the impact of pulsed magnetic therapy, aerobic exercise, and a combination of both modalities on osteoporotic female patients postthyroidectomy. METHODS Between May 2018 and September 2019, 45 female patients with osteoporosis were included in the randomized clinical study, their age ranged from 40 to 50 years, had thyroidectomy for at least 6 months ago, and had an inactive lifestyle for at least the previous 6 months. Patients were assigned randomly into 3 equal groups. Group A (magnetic therapy group): received routine medical treatment (bisphosphonates, calcium, and vitamin D) in addition to pulsed magnetic therapy on the hip region for 12 weeks (3 sessions/week). Group B (exercise group): received routine medical treatment plus moderate-intensity aerobic exercise for 12 weeks (3 sessions/week). Group C (combined magnetic therapy and exercise therapy group): received routine medical treatment plus pulsed magnetic therapy and moderate-intensity aerobic exercise for 12 weeks (3 sessions/week). The 3 groups were assessed for bone mineral density (BMD) at baseline by dual-energy x-ray absorptiometry and after 12 weeks of treatment. RESULTS The results showed that within-group analysis a statistically significant increase was reveled (P < .05) for BMD in the 3 studied groups. Comparing the results among the 3 tested groups revealed a significant increase (P < .05) in posttesting mean values of BMD in group (C) compared to group (A) and group (B). No significant statistical difference in BMD means values between the 2 groups (A) and (B) after testing was detected. CONCLUSION Combination of both pulsed magnetic therapy and moderate-intensity aerobic exercise showed significant improvement in BMD at the hip region than using any of the 2 modalities alone.
Collapse
Affiliation(s)
- Marwa M. Eid
- Department of Physical Therapy, College of Applied Medical Sciences, Taif University, Taif, Saudi Arabia
- Department of Physical Therapy for Surgery, Faculty of Physical Therapy, Cairo University, Giza, Egypt
| | - Amira M. El-Gendy
- Department of Physical Therapy, College of Applied Medical Sciences, Taif University, Taif, Saudi Arabia
- Department of Basic Sciences, Faculty of Physical Therapy, Cairo University, Giza, Egypt
| | - Walid Kamal Abdelbasset
- Department of Health and Rehabilitation Sciences, College of Applied Medical Sciences, Prince Sattam bin Abdulaziz University, Al-Kharj, Saudi Arabia
- Department of Physical Therapy, Kasr Al-Aini Hospital, Cairo University, Giza, Egypt
| | - Safaa Mostafa Elkholi
- Department of Rehabilitation Sciences, Faculty of Health and Rehabilitation Sciences, Princess Nourah bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Mostafa S. Abdel-fattah
- Department of Physical Therapy for Cardiovascular/Respiratory Disorder and Geriatrics, Faculty of Physical Therapy, Cairo University, Giza, Egypt
| |
Collapse
|
20
|
Wu M, Zou L, Jiang L, Zhao Z, Liu J. Osteoinductive and antimicrobial mechanisms of graphene-based materials for enhancing bone tissue engineering. J Tissue Eng Regen Med 2021; 15:915-935. [PMID: 34469046 DOI: 10.1002/term.3239] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 07/23/2021] [Accepted: 08/03/2021] [Indexed: 02/05/2023]
Abstract
Graphene-based materials (GMs) have great application prospects in bone tissue engineering due to their osteoinductive ability and antimicrobial activity. GMs induce osteogenic differentiation through several mechanisms and pathways in bone tissue engineering. First of all, the surface and high hardness of the porous folds of graphene or graphene oxide (GO) can generate mechanical stimulation to initiate a cascade of reactions that promote osteogenic differentiation without any chemical inducers. In addition, change of the extracellular matrix (ECM), regulation of macrophage polarization, the oncostatin M (OSM) signaling pathway, the MAPK signaling pathway, the BMP signaling pathway, the Wnt/β-catenin signaling pathway, and other pathways are involved in GMs' regulation of osteogenesis. In bone tissue engineering, GMs prevent the formation of microbial biofilms mainly through preventing microbial adhesion and killing them. The former is mainly achieved by reducing surface free energy (SFE) and increasing hydrophobicity. The latter mainly includes oxidative stress and photothermal/photodynamic effects. Graphene and its derivatives (GDs) are mainly combined with bioactive ceramic materials, metal materials and macromolecular polymers to play an antimicrobial effect in bone tissue engineering. Concentration, number of layers, and type of GDs often affect the antimicrobial activity of GMs. In this paper, we reviewed relevant osteoinductive and antimicrobial mechanisms of GMs and their applications in bone tissue engineering.
Collapse
Affiliation(s)
- Mengsong Wu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Ling Zou
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Department of Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Linli Jiang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Zhihe Zhao
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Jun Liu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
21
|
Del Buono A, Zampogna B, Osti L, Fontanarosa A, Garofalo R, Papalia R. Pulsed electromagnetic fields after intramedullary nailing of tibial fractures: a case control study. INTERNATIONAL ORTHOPAEDICS 2021; 45:2945-2950. [PMID: 34448925 DOI: 10.1007/s00264-021-05125-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Accepted: 06/20/2021] [Indexed: 12/26/2022]
Abstract
PURPOSE To compare clinical and functional outcomes of two groups of patients undergoing reduction and nailing fixation for diaphyseal fractures of the tibia with (PEMF group) and without (control group) post-operative pulsed electromagnetic field (PEMF) application. METHODS This is a retrospective study on 50 patients (mean age 43.3 years, 28 males and 22 females) with diaphyseal tibial fractures managed between 2017 and 2019. Twenty-five patients underwent reduction, nailing fixation, and PEMF application post-operatively (PEMF group) and 25 patients underwent nailing fixation. Radiographic imaging assessment was performed every month until fracture healing had been evident. Use of analgesics, fracture healing time, post-operative lower limb alignment, and post-operative complications were recorded. Patients were asked about return to preinjury activity. All patients were assessed at 3 months and at an average follow-up of 13 months. The VAS scale and Johner-Wruhs criteria were used for pain assessment and functional recovery, respectively. RESULTS Comparing groups, VAS values were significantly lower in the PEMF group at three months and comparable at one year. The patients in the PEMF group took an average of 4.1 months to resume their preinjury activities, and control patients took an average of 5.3 months (P < 0.0001). According to the Johner-Wruhs score, the effective rate was 100% (25/25) in the PEMF group and 92% (23/25) in the control group (P = 0.14). CONCLUSIONS PEMF application after intramedullary nailing is safe and reduces post-operative pain, use of analgesics, and the time of healing fracture. At one year, there is no difference in outcome measures, regardless of PEMF application.
Collapse
Affiliation(s)
- Angelo Del Buono
- Unit of Orthopedics and Trauma Surgery, L. Curto Hospital, Polla, Italy
| | - Biagio Zampogna
- Department of Orthopedics and Trauma Surgery, Campus Bio-Medico University of Rome, Via Alvaro del Portillo, 200, 00128, Rome, Italy
| | - Leonardo Osti
- Unit of Arthroscopy and Sports Medicine, Hesperia Hospital, Modena, Italy
| | - Alberto Fontanarosa
- Department of Shoulder Surgery and Sport Trauma Surgery, F. Miulli Hospital, Acquaviva Delle Fonti, BA, Italy
| | - Raffaele Garofalo
- Department of Shoulder Surgery and Sport Trauma Surgery, F. Miulli Hospital, Acquaviva Delle Fonti, BA, Italy
| | - Rocco Papalia
- Department of Orthopedics and Trauma Surgery, Campus Bio-Medico University of Rome, Via Alvaro del Portillo, 200, 00128, Rome, Italy.
| |
Collapse
|
22
|
Epigenetic dysregulation in various types of cells exposed to extremely low-frequency magnetic fields. Cell Tissue Res 2021; 386:1-15. [PMID: 34287715 PMCID: PMC8526474 DOI: 10.1007/s00441-021-03489-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Accepted: 06/18/2021] [Indexed: 02/07/2023]
Abstract
Epigenetic mechanisms regulate gene expression, without changing the DNA sequence, and establish cell-type-specific temporal and spatial expression patterns. Alterations of epigenetic marks have been observed in several pathological conditions, including cancer and neurological disorders. Emerging evidence indicates that a variety of environmental factors may cause epigenetic alterations and eventually influence disease risks. Humans are increasingly exposed to extremely low-frequency magnetic fields (ELF-MFs), which in 2002 were classified as possible carcinogens by the International Agency for Research on Cancer. This review summarizes the current knowledge of the link between the exposure to ELF-MFs and epigenetic alterations in various cell types. In spite of the limited number of publications, available evidence indicates that ELF-MF exposure can be associated with epigenetic changes, including DNA methylation, modifications of histones and microRNA expression. Further research is needed to investigate the molecular mechanisms underlying the observed phenomena.
Collapse
|
23
|
Pulsed Electromagnetic Fields Modulate miRNAs During Osteogenic Differentiation of Bone Mesenchymal Stem Cells: a Possible Role in the Osteogenic-angiogenic Coupling. Stem Cell Rev Rep 2021; 16:1005-1012. [PMID: 32681233 DOI: 10.1007/s12015-020-10009-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Despite the high intrinsic ability of bone tissue to regenerate, bone healing fails in some pathological conditions and especially in the presence of large defects. Due to the strong relationship between bone development and vascularization during in vivo bone formation and repair, strategies promoting the osteogenic-angiogenic coupling are crucial for regenerative medicine. Increasing evidence shows that miRNAs play important roles in controlling osteogenesis and bone vascularization and are important tool in medical research although their clinical use still needs to optimize miRNA stability and delivery. Pulsed electromagnetic fields (PEMFs) have been successfully used to enhance bone repair and their clinical activity has been associated to their ability to promote the osteogenic differentiation of human mesenchymal stem cells (hMSCs). In this study we investigated the potential ability of PEMF exposure to modulate selected miRNAs involved in the osteogenic differentiation of human bone mesenchymal stem cells (hBMSCs). We show that, during in vitro hBMSC differentiation, PEMFs up-modulate the expression of miR-26a and miR-29b, which favor osteogenic differentiation, and decrease miR-125b which acts as an inhibitor miRNA. As PEMFs promote the expression and release of miRNAs also involved in angiogenesis, we conclude that PEMFs may represent a noninvasive and safe strategy to modulate miRNAs with relevant roles in bone repair and with the potential to regulate the osteogenic-angiogenic coupling.
Collapse
|
24
|
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: 0] [Impact Index Per Article: 0] [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
|
25
|
Pulsed Electromagnetic Field Stimulation in Osteogenesis and Chondrogenesis: Signaling Pathways and Therapeutic Implications. Int J Mol Sci 2021; 22:ijms22020809. [PMID: 33467447 PMCID: PMC7830993 DOI: 10.3390/ijms22020809] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 12/30/2020] [Accepted: 01/11/2021] [Indexed: 02/07/2023] Open
Abstract
Mesenchymal stem cells (MSCs) are the main cell players in tissue repair and thanks to their self-renewal and multi-lineage differentiation capabilities, they gained significant attention as cell source for tissue engineering (TE) approaches aimed at restoring bone and cartilage defects. Despite significant progress, their therapeutic application remains debated: the TE construct often fails to completely restore the biomechanical properties of the native tissue, leading to poor clinical outcomes in the long term. Pulsed electromagnetic fields (PEMFs) are currently used as a safe and non-invasive treatment to enhance bone healing and to provide joint protection. PEMFs enhance both osteogenic and chondrogenic differentiation of MSCs. Here, we provide extensive review of the signaling pathways modulated by PEMFs during MSCs osteogenic and chondrogenic differentiation. Particular attention has been given to the PEMF-mediated activation of the adenosine signaling and their regulation of the inflammatory response as key player in TE approaches. Overall, the application of PEMFs in tissue repair is foreseen: (1) in vitro: to improve the functional and mechanical properties of the engineered construct; (2) in vivo: (i) to favor graft integration, (ii) to control the local inflammatory response, and (iii) to foster tissue repair from both implanted and resident MSCs cells.
Collapse
|
26
|
Alekseeva LI, Byalovsky YY, Zagorodny NV, Ivanova GE, Karateev DE, Konchugova TV, Rakitina IS, Strakhov MA. [Pathophysiological mechanisms of the therapeutic action of alternating electromagnetic fields in the treatment of osteoarticular pathology]. VOPROSY KURORTOLOGII, FIZIOTERAPII, I LECHEBNOI FIZICHESKOI KULTURY 2021; 98:80-90. [PMID: 34223758 DOI: 10.17116/kurort20219803180] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Treatment of osteoarticular pathology with an alternating electromagnetic field (AEMF) is used today as a promising, non-invasive and safe strategy of physiotherapy. It has been shown that the action of alternating electromagnetic fields on the musculoskeletal system triggers signaling cascades that effectively contribute to the restoration of bone and articular tissue. The pathophysiological mechanisms underlying the cellular and subcellular effects of stimulation by an alternating electromagnetic field during the restoration of bone and articular tissue are considered. It was pointed out the several key signaling pathways involved in the restoration of bone and articular tissue under the influence of electromagnetic fields with an analysis of the potential for therapeutic application of electromagnetic fields alone or in combination with other available therapies.
Collapse
Affiliation(s)
- L I Alekseeva
- V.A. Nasonova Research Institute of Rheumatology, Moscow, Russia
| | | | - N V Zagorodny
- N.N. Priorov Central Research Institute of Traumatology and Orthopedics, Moscow, Russia
| | - G E Ivanova
- N.I. Pirogov Russian National Research Medical University, Moscow, Russia
| | - D E Karateev
- M.F. Vladimirsky Moscow Regional Research and Clinical Institute, Moscow, Russia
| | - T V Konchugova
- National Medical Research Center for Rehabilitation and Balneology, Moscow, Russia
- I.M. Sechenov First Moscow State Medical University, Moscow, Russia
| | | | - M A Strakhov
- N.I. Pirogov Russian National Research Medical University, Moscow, Russia
| |
Collapse
|
27
|
Kurniawan A, Kodrat E, Gani YI. Effectiveness of granulocyte colony stimulating factor to enhance healing on delayed union fracture model Sprague-Dawley rat. Ann Med Surg (Lond) 2021; 61:54-60. [PMID: 33384875 PMCID: PMC7770509 DOI: 10.1016/j.amsu.2020.12.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 12/01/2020] [Accepted: 12/02/2020] [Indexed: 01/08/2023] Open
Abstract
INTRODUCTION Delayed union is a problem that can occur after fracture healing. Many studies were conducted based on the diamond concept approach to solve the problem of delayed union. Granulocyte-colony stimulating factor (G-CSF) is one of the various substances known to have a positive role in healing skeletal tissue or adjuvant regeneration. This study was conducted to see the effect of G-CSF in affecting delayed union fracture healing. MATERIALS AND METHOD The experimental study was conducted by randomized posttest only control group design on 24 experimental animals Sprague-Dawley white rats that had experienced delayed union models. The study compared the treatment group injected with subcutaneous G-CSF with a control group and was divided into four groups (n = 6). Harvest and follow-up histomorphometry and immunohistochemistry were performed in the second week and in the fourth week the histomorphometry analysis consisted of the percentage of immature bone area, cartilage, and fibrous area. The semiquantitative evaluation of immunohistochemistry with the expression of BMP-2 through the immunoreactive score (IRS). RESULT In the evaluation of histomorphometry and immunohistochemical parameters, there were significantly more woven bone area (p = 0,015), less fibrosis area (p = 0,002) and higher BMP 2 expression (p = 0,004) in treatment group week four compared to control. . CONCLUSION G-CSF was shown to increase the speed of healing in Sprague-Dawley rats on delayed union models evaluated from histomorphometry and immunohistochemical aspects.
Collapse
Affiliation(s)
- Aryadi Kurniawan
- Paediatric Orthopaedic Division, Department of Orthopaedics and Traumatology, Faculty of Medicine Universitas Indonesia-Cipto Mangunkusumo Hospital, Jakarta, Indonesia
| | - Evelina Kodrat
- Musculoskletal Pathology Division, Departement of Anatomic Phatology, Faculty of Medicine Universitas Indonesia-Cipto Mangunkusumo Hospital, Jakarta, Indonesia
| | - Yogi Ismail Gani
- Orthopaedics and Traumatology, Faculty of Medicine, Universitas Indonesia-Cipto Mangunkusumo Hospital, Jakarta, Indonesia
| |
Collapse
|
28
|
Zhang Z, Zheng T, Zhu R. Microchip with Single-Cell Impedance Measurements for Monitoring Osteogenic Differentiation of Mesenchymal Stem Cells under Electrical Stimulation. Anal Chem 2020; 92:12579-12587. [PMID: 32859132 DOI: 10.1021/acs.analchem.0c02556] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Effective induction methods and in situ monitoring are essential for studying the mechanism of biological responses in stem cell differentiation. This article proposes an induction method incorporating electrical stimulation under an inhomogeneous field with single-cell impedance monitoring for studying osteogenic differentiation of mesenchymal stem cells (MSCs) using a microchip. The microchip contains an array of sextupole-electrode units for implementing a combination of controllable electrical stimulation and single-cell impedance measurements. MSCs are inducted to osteogenic differentiation under electrical stimulation using quadrupole electrodes and single-cell impedances are monitored in situ using a pair of microelectrodes at each unit center. The proposed microchip adopts an array design to monitor a number of MSCs in parallel, which improves measurement throughput and facilitates to carry out statistic tests. We perform osteogenic differentiation of MSCs on the microchip with and without electrical stimulation meanwhile monitoring single-cell impedance in real time for 21 days. The recorded impedance results show the detailed characteristic change of MSCs at the single-cell level during osteogenic differentiation, which demonstrates a significant difference between the conditions with and without electrical stimulation. The cell morphology and various staining analyses are also used to validate osteogenesis and correlate with the impedance expression. Correlation analysis of the impedance measurement, cell morphology, and various staining assays proves the great acceleration effect of the proposed electrical stimulation on osteogenic differentiation of MSCs. The proposed impedance method can monitor the dynamic process of cell development and study heterogeneity of stem cell differentiation at the single-cell level.
Collapse
Affiliation(s)
- Zhizhong Zhang
- State Key Laboratory of Precision Measurements Technology and Instruments, Department of Precision Instrument, Tsinghua University, Beijing 100084, China
| | - Tianyang Zheng
- State Key Laboratory of Precision Measurements Technology and Instruments, Department of Precision Instrument, Tsinghua University, Beijing 100084, China
| | - Rong Zhu
- State Key Laboratory of Precision Measurements Technology and Instruments, Department of Precision Instrument, Tsinghua University, Beijing 100084, China
| |
Collapse
|
29
|
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: 24] [Impact Index Per Article: 6.0] [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
|
30
|
Lullini G, Cammisa E, Setti S, Sassoli I, Zaffagnini S, Marcheggiani Muccioli GM. Role of pulsed electromagnetic fields after joint replacements. World J Orthop 2020; 11:285-293. [PMID: 32572365 PMCID: PMC7298453 DOI: 10.5312/wjo.v11.i6.285] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2020] [Revised: 05/14/2020] [Accepted: 05/28/2020] [Indexed: 02/06/2023] Open
Abstract
Although the rate of patients reporting satisfaction is generally high after joint replacement surgery, up to 23% after total hip replacement and 34% after total knee arthroplasty of treated subjects report discomfort or pain 1 year after surgery. Moreover, chronic or subacute inflammation is reported in some cases even a long time after surgery. Another open and debated issue in prosthetic surgery is implant survivorship, especially when related to good prosthesis bone ingrowth. Pulsed Electro Magnetic Fields (PEMFs) treatment, although initially recommended after total joint replacement to promote bone ingrowth and to reduce inflammation and pain, is not currently part of usual clinical practice. The purpose of this review was to analyze existing literature on PEMFs effects in joint replacement surgery and to report results of clinical studies and current indications. We selected all currently available prospective studies or RCT on the use of PEMFs in total joint replacement with the purpose of investigating effects of PEMFs on recovery, pain relief and patients’ satisfaction following hip, knee or shoulder arthroplasty. All the studies analyzed reported no adverse effects, and good patient compliance to the treatment. The available literature shows that early control of joint inflammation process in the first days after surgery through the use of PEMFs should be considered an effective completion of the surgical procedure to improve the patient’s functional recovery.
Collapse
Affiliation(s)
- Giada Lullini
- Laboratorio di Analisi del Movimento e di valutazione funzionale protesi, IRCCS Istituto Ortopedico Rizzoli - DIBINEM - University of Bologna, Bologna 40100, Italy
| | - Eugenio Cammisa
- II Orthopaedic and Traumatology Clinic, IRCCS Istituto Ortopedico Rizzoli - DIBINEM - University of Bologna, Bologna 40100, Italy
| | - Stefania Setti
- Laboratory of Clinical Biophysics, IGEA S.p.A. Clinical Biophysics, 41012 Carpi (Mo), Italy
| | - Iacopo Sassoli
- II Orthopaedic and Traumatology Clinic, IRCCS Istituto Ortopedico Rizzoli - DIBINEM - University of Bologna, Bologna 40100, Italy
| | - Stefano Zaffagnini
- II Orthopaedic and Traumatology Clinic, IRCCS Istituto Ortopedico Rizzoli - DIBINEM - University of Bologna, Bologna 40100, Italy
| | | |
Collapse
|
31
|
Pulsed Electromagnetic Field Stimulation of Bone Healing and Joint Preservation: Cellular Mechanisms of Skeletal Response. JOURNAL OF THE AMERICAN ACADEMY OF ORTHOPAEDIC SURGEONS GLOBAL RESEARCH AND REVIEWS 2020; 4:e1900155. [PMID: 33970582 PMCID: PMC7434032 DOI: 10.5435/jaaosglobal-d-19-00155] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The US FDA has approved pulsed electromagnetic fields (PEMFs) as a safe and effective treatment for nonunions of bone. Despite its clinical use, the mechanisms of action of electromagnetic stimulation of the skeleton have been elusive. Recently, cell membrane receptors have been identified as the site of action of PEMF and provide a mechanistic rationale for clinical use. This review highlights key processes in cell responses to PEMF as follows: (1) signal transduction through A2A and A3 adenosine cell membrane receptors and (2) dose-response effects on the synthesis of structural and signaling extracellular matrix (ECM) components. Through these actions, PEMF can increase the structural integrity of bone and cartilage ECM, enhancing repair, and alter the homeostatic balance of signaling cytokines, producing anti-inflammatory effects. PEMFs exert a proanabolic effect on the bone and cartilage matrix and a chondroprotective effect counteracting the catabolic effects of inflammation in the joint environment. Understanding of PEMF membrane targets, and of the specific intracellular pathways involved, culminating in the synthesis of ECM proteins and reduction in inflammatory cytokines, should enhance confidence in the clinical use of PEMF and the identification of clinical conditions likely to be affected by PEMF exposure.
Collapse
|
32
|
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: 103] [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
|
33
|
Bone Morphogenetic Protein-2 Signaling in the Osteogenic Differentiation of Human Bone Marrow Mesenchymal Stem Cells Induced by Pulsed Electromagnetic Fields. Int J Mol Sci 2020; 21:ijms21062104. [PMID: 32204349 PMCID: PMC7139765 DOI: 10.3390/ijms21062104] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Revised: 03/16/2020] [Accepted: 03/17/2020] [Indexed: 12/12/2022] Open
Abstract
Pulsed electromagnetic fields (PEMFs) are clinically used with beneficial effects in the treatment of bone fracture healing. This is due to PEMF ability to favor the osteogenic differentiation of mesenchymal stem cells (MSCs). Previous studies suggest that PEMFs enhance the osteogenic activity of bone morphogenetic protein-2 (BMP2) which is used in various therapeutic interventions. This study investigated the molecular events associated to the synergistic activity of PEMFs and BMP2 on osteogenic differentiation. To this aim, human MSCs (hMSCs) were exposed to PEMFs (75 Hz, 1.5 mT) in combination with BMP2, upon detection of the minimal dose able to induce differentiation. Changes in the expression of BMP signaling pathway genes including receptors and ligands, as well as in the phosphorylation of BMP downstream signaling proteins, such as SMAD1/5/8 and MAPK, were analyzed. Results showed the synergistic activity of PEMFs and BMP2 on osteogenic differentiation transcription factors and markers. The PEMF effects were associated to the increase in BMP2, BMP6, and BMP type I receptor gene expression, as well as SMAD1/5/8 and p38 MAPK activation. These results increase knowledge concerning the molecular events involved in PEMF stimulation showing that PEMFs favor hMSCs osteogenic differentiation by the modulation of BMP signaling components.
Collapse
|
34
|
Zhang B, Xie Y, Ni Z, Chen L. Effects and Mechanisms of Exogenous Electromagnetic Field on Bone Cells: A Review. Bioelectromagnetics 2020; 41:263-278. [PMID: 32159242 DOI: 10.1002/bem.22258] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Accepted: 02/25/2020] [Indexed: 12/16/2022]
Abstract
Osteoporosis, fractures, and other bone diseases or injuries represent serious health problems in modern society. A variety of treatments including drugs, surgeries, physical therapies, etc. have been used to prevent or delay the progression of these diseases/injuries with limited effects. Electromagnetic field (EMF) has been used to non-invasively treat bone diseases, such as fracture and osteoporosis, for many years. However, because a variety of cellular and molecular events can be affected by EMF with various parameters, the precise bioeffects and underlying mechanisms of specific EMF on bone cells are still obscure. Here, we summarize the common therapeutic parameters (frequency and intensity) of major types of EMF used to treat bone cells taken from 32 papers we selected from the PubMed database published in English from 1991 to 2018. Briefly, pulse EMF promotes the proliferation of osteoblasts when its frequency is 7.5-15 Hz or 50-75 Hz and the intensity is 0.40-1.55 mT or 3.8-4 mT. Sinusoidal EMF, with 0.9-4.8 mT and 45-60 Hz, and static magnetic field with 0.1-0.4 mT or 400 mT, can promote osteoblast differentiation and maturation. Finally, we summarize the latest advances on the molecular signaling pathways influenced by EMF in osteoblasts and osteoclasts. A variety of molecules such as adenosine receptors, calcium channels, BMP2, Notch, Wnt1, etc., can be influenced by EMF in osteoblasts. For osteoclasts, EMF affects RANK, NF-κB, MAPK, etc. We speculate that EMF with different frequencies and intensities exert distinct bioeffects on specific bone cells. More high-quality work is required to explore the detailed effects and underlying mechanisms of EMF on bone cells/skeleton to optimize the application of EMF on bone diseases/injuries. Bioelectromagnetics. 2020;41:263-278 © 2020 Bioelectromagnetics Society.
Collapse
Affiliation(s)
- Bin Zhang
- State Key Laboratory of Trauma, Burns and Combined Injury, Department of Wound Repair and Rehabilitation, Center of Bone Metabolism and Repair, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China.,Center of Rehabilitation, Xingcheng Sanatorium of PLA Strategic Support Force, Xingcheng, China
| | - Yangli Xie
- State Key Laboratory of Trauma, Burns and Combined Injury, Department of Wound Repair and Rehabilitation, Center of Bone Metabolism and Repair, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Zhenhong Ni
- State Key Laboratory of Trauma, Burns and Combined Injury, Department of Wound Repair and Rehabilitation, Center of Bone Metabolism and Repair, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Lin Chen
- State Key Laboratory of Trauma, Burns and Combined Injury, Department of Wound Repair and Rehabilitation, Center of Bone Metabolism and Repair, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| |
Collapse
|
35
|
Fan C, Zhan SH, Dong ZX, Yang W, Deng WS, Liu X, Wang DA, Sun P. Cross-linked gelatin microsphere-based scaffolds as a delivery vehicle of MC3T3-E1 cells: in vitro and in vivo evaluation. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2020; 108:110399. [DOI: 10.1016/j.msec.2019.110399] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Revised: 10/29/2019] [Accepted: 11/04/2019] [Indexed: 12/21/2022]
|
36
|
Zheng T, Zhang Z, Zhu R, Sun D. A microelectrode array chip for osteogenic differentiation of mesenchymal stem cells under electrical stimulation. LAB ON A CHIP 2020; 20:373-383. [PMID: 31850469 DOI: 10.1039/c9lc01081e] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Electrical stimulation (ES) as an easy and effective inducing method has been widely used in induction differentiation of stem cells, e.g. osteogenic differentiation of mesenchymal stem cells (MSCs) for bone healing and bone tissue therapies. However, the micro-effect of an inhomogeneous electric field has rarely been investigated for ES in induction differentiation, and conventionally used ex situ assays may preclude accurate assessment due to variation from cell inoculation and treatments. Here, a novel electrical stimulation method with a microelectrode array chip is proposed for osteogenic differentiation of MSCs. The electric field applied onto the MSCs by the microelectrode array is designed similarly with a natural aggregation distribution of differentiated MSCs. The proposed ES method accelerates osteoblast proliferation and differentiation in the electrode array region and generates a larger amount of mineralized deposits, which are assayed via in situ alizarin red staining and morphology observation as well as immunocytochemistry. In addition, this method allows a direct in situ assessment to compare the osteogenic differentiation of MSCs with and without ES on a single chip to avoid culture environment difference. The method provides a fundamental platform for investigating induced differentiation of stem cells and allows integration with multifunctional cell assays to achieve in situ tracking for the differentiation process of stem cells.
Collapse
Affiliation(s)
- Tianyang Zheng
- State Key Laboratory of Precision Measurement Technology and Instruments, Department of Precision Instrument, Tsinghua University, Beijing, 100084, China.
| | - Zhizhong Zhang
- State Key Laboratory of Precision Measurement Technology and Instruments, Department of Precision Instrument, Tsinghua University, Beijing, 100084, China.
| | - Rong Zhu
- State Key Laboratory of Precision Measurement Technology and Instruments, Department of Precision Instrument, Tsinghua University, Beijing, 100084, China.
| | - Dong Sun
- Department of Mechanical and Biomedical Engineering, City University of Hong Kong, Hong Kong, China
| |
Collapse
|
37
|
Nasb M, Liangjiang H, Gong C, Hong C. Human adipose-derived Mesenchymal stem cells, low-intensity pulsed ultrasound, or their combination for the treatment of knee osteoarthritis: study protocol for a first-in-man randomized controlled trial. BMC Musculoskelet Disord 2020; 21:33. [PMID: 31941483 PMCID: PMC6964002 DOI: 10.1186/s12891-020-3056-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2019] [Accepted: 01/08/2020] [Indexed: 01/22/2023] Open
Abstract
Background Human adipose-derived Mesenchymal stem cells (HADMSCs) have proven their efficacy in treating osteoarthritis (OA), in earlier preclinical and clinical studies. As the tissue repairers are under the control of mechanical and biochemical signals, improving regeneration outcomes using such signals has of late been the focus of attention. Among mechanical stimuli, low-intensity pulsed ultrasound (LIPUS) has recently shown promise both in vitro and in vivo. This study will investigate the potential of LIPUS in enhancing the regeneration process of an osteoarthritic knee joint. Methods This study involves a prospective, randomized, placebo-controlled, and single-blind trial based on the SPIRIT guidelines, and aims to recruit 96 patients initially diagnosed with knee osteoarthritis, following American College of Rheumatology criteria. Patients will be randomized in a 1:1:1 ratio to receive Intraarticular HADMSCs injection with LIPUS, Intraarticular HADMSCs injection with shame LIPUS, or Normal saline with LIPUS. The primary outcome is Western Ontario and McMaster Universities Index of OA (WOMAC) score, while the secondary outcomes will be other knee structural changes, and lower limb muscle strength such as the knee cartilage thickness measured by MRI. Blinded assessments will be performed at baseline (1 month prior to treatment), 1 month, 3 months, and 6 months following the interventions. Discussion This trial will be the first clinical study to comprehensively investigate the safety and efficacy of LIPUS on stem cell therapy in OA patients. The results may provide evidence of the effectiveness of LIPUS in improving stem cell therapy and deliver valuable information for the design of subsequent trials. Trial registration This study had been prospectively registered with the Chinese Clinical Trials Registry. registration number: ChiCTR1900025907 at September 14, 2019.
Collapse
Affiliation(s)
- Mohammad Nasb
- Department of Rehabilitation Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China.,Department of Physical Therapy, Health science faculty, Albaath University, Homs, Syria
| | - Huang Liangjiang
- Department of Rehabilitation Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China
| | - Chenzi Gong
- Department of Rehabilitation Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China
| | - Chen Hong
- Department of Rehabilitation Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China.
| |
Collapse
|
38
|
Oliveira KMC, Barker JH, Berezikov E, Pindur L, Kynigopoulos S, Eischen-Loges M, Han Z, Bhavsar MB, Henrich D, Leppik L. Electrical stimulation shifts healing/scarring towards regeneration in a rat limb amputation model. Sci Rep 2019; 9:11433. [PMID: 31391536 PMCID: PMC6685943 DOI: 10.1038/s41598-019-47389-w] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Accepted: 07/16/2019] [Indexed: 12/19/2022] Open
Abstract
Different species respond differently to severe injury, such as limb loss. In species that regenerate, limb loss is met with complete restoration of the limbs’ form and function, whereas in mammals the amputated limb’s stump heals and scars. In in vitro studies, electrical stimulation (EStim) has been shown to promote cell migration, and osteo- and chondrogenesis. In in vivo studies, after limb amputation, EStim causes significant new bone, cartilage and vessel growth. Here, in a rat model, the stumps of amputated rat limbs were exposed to EStim, and we measured extracellular matrix (ECM) deposition, macrophage distribution, cell proliferation and gene expression changes at early (3 and 7 days) and later stages (28 days). We found that EStim caused differences in ECM deposition, with less condensed collagen fibrils, and modified macrophage response by changing M1 to M2 macrophage ratio. The number of proliferating cells was increased in EStim treated stumps 7 days after amputation, and transcriptome data strongly supported our histological findings, with activated gene pathways known to play key roles in embryonic development and regeneration. In conclusion, our findings support the hypothesis that EStim shifts injury response from healing/scarring towards regeneration. A better understanding of if and how EStim controls these changes, could lead to strategies that replace scarring with regeneration.
Collapse
Affiliation(s)
- K M C Oliveira
- Frankfurt Initiative for Regenerative Medicine, Experimental Orthopedics & Trauma Surgery, J.W. Goethe University, Frankfurt am Main, Germany
| | - J H Barker
- Frankfurt Initiative for Regenerative Medicine, Experimental Orthopedics & Trauma Surgery, J.W. Goethe University, Frankfurt am Main, Germany
| | - E Berezikov
- European Research Institute for the Biology of Ageing, University Medical Center Groningen, Groningen, The Netherlands
| | - L Pindur
- Frankfurt Initiative for Regenerative Medicine, Experimental Orthopedics & Trauma Surgery, J.W. Goethe University, Frankfurt am Main, Germany.,Department of Plastic, Hand and Reconstructive Surgery, BG Trauma Center Frankfurt am Main gGmbH, Frankfurt am Main, Germany
| | - S Kynigopoulos
- Frankfurt Initiative for Regenerative Medicine, Experimental Orthopedics & Trauma Surgery, J.W. Goethe University, Frankfurt am Main, Germany
| | - M Eischen-Loges
- Frankfurt Initiative for Regenerative Medicine, Experimental Orthopedics & Trauma Surgery, J.W. Goethe University, Frankfurt am Main, Germany
| | - Z Han
- Frankfurt Initiative for Regenerative Medicine, Experimental Orthopedics & Trauma Surgery, J.W. Goethe University, Frankfurt am Main, Germany
| | - M B Bhavsar
- Frankfurt Initiative for Regenerative Medicine, Experimental Orthopedics & Trauma Surgery, J.W. Goethe University, Frankfurt am Main, Germany
| | - D Henrich
- Department of Trauma, Hand and Reconstructive Surgery, J.W. Goethe University, Frankfurt am Main, Germany
| | - L Leppik
- Frankfurt Initiative for Regenerative Medicine, Experimental Orthopedics & Trauma Surgery, J.W. Goethe University, Frankfurt am Main, Germany.
| |
Collapse
|
39
|
Abstract
Non-union of bone following fracture is an orthopaedic condition with a high morbidity and clinical burden. Despite its estimated global prevalence of nine million annually, the limit of bone regeneration therapy still results in patients living with pain, a reduced quality of life and associated psychological, social and financial repercussions. This review provides an overview of the current epidemiological and aetiological data, and highlights where the clinical challenges in treating non-union lie. Current treatment strategies are discussed as well as promising future research foci. Development in biotechnologies to treat non-union provides exciting scope for more effective treatment for this debilitating condition.
Collapse
Affiliation(s)
- S K Stewart
- Department of Bioengineering, Imperial College London, United Kingdom
| |
Collapse
|
40
|
Huang Y, Deng H, Fan Y, Zheng L, Che J, Li X, Aifantis KE. Conductive nanostructured Si biomaterials enhance osteogeneration through electrical stimulation. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2019; 103:109748. [PMID: 31349398 DOI: 10.1016/j.msec.2019.109748] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Revised: 04/05/2019] [Accepted: 05/12/2019] [Indexed: 02/07/2023]
Abstract
It is well known that the differentiation of stem cells is affected by the cell culture medium, the scaffold surface and electrochemical signals. However, stimulation of patterned biomaterials seeded with stem cell cultures has not been explored. Herein the effect of electrical stimulation on osteogenic differentiation of rat bone marrow-derived mesenchymal stem cells (rBMSCs) cultured on solid and nanoporous micropyramid patterned Si surfaces was evaluated. It was found that both stimulation and scaffold patterning significantly enhanced osteo-differentiation. The stimulated nanoporous micropyramid scaffolds were more promising compared to the stimulated solid micropyramid surfaces, as they significantly promoted the osteogenic differentiation of rBMSCs via BMP/Smad signaling pathway. Particularly, as compared to the unstimulated patterned biomaterials, the stimulated patterned scaffolds allowed for a significant increase in core binding factor alpha l, alkaline phosphatase, the alpha l chain of type I Col, osteocalcin, and osteonectin, all of which are characteristic for osteo-differentiation. The proposed combination of electrical stimulation with scaffold patterning may provide novel promising strategies for bone tissue engineering and regenerative medicine.
Collapse
Affiliation(s)
- Yan Huang
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, China; Beijing Advanced Innovation Center for Biomedical Engineering, Beihang University, Beijing 100083, China
| | | | - Yubo Fan
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, China; Beijing Advanced Innovation Center for Biomedical Engineering, Beihang University, Beijing 100083, China; Beijing Key Laboratory of Rehabilitation Technical Aids for Old-Age Disability, National Research Center for Rehabilitation Technical Aids, Beijing 100176, China
| | - Lisha Zheng
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, China; Beijing Advanced Innovation Center for Biomedical Engineering, Beihang University, Beijing 100083, China
| | - Jifei Che
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, China
| | - Xiaoming Li
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, China; Beijing Advanced Innovation Center for Biomedical Engineering, Beihang University, Beijing 100083, China.
| | - Katerina E Aifantis
- Department of Mechanical and Aerospace Engineering, University of Florida, Gainesville, FL 32611, USA.
| |
Collapse
|
41
|
Wang YY, Pu XY, Shi WG, Fang QQ, Chen XR, Xi HR, Gao YH, Zhou J, Xian CJ, Chen KM. Pulsed electromagnetic fields promote bone formation by activating the sAC-cAMP-PKA-CREB signaling pathway. J Cell Physiol 2019; 234:2807-2821. [PMID: 30067871 DOI: 10.1002/jcp.27098] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2018] [Accepted: 06/28/2018] [Indexed: 02/06/2023]
Abstract
The application of pulsed electromagnetic fields (PEMFs) in the prevention and treatment of osteoporosis has long been an area of interest. However, the clinical application of PEMFs remains limited because of the poor understanding of the PEMF action mechanism. Here, we report that PEMFs promote bone formation by activating soluble adenylyl cyclase (sAC), cyclic adenosine monophosphate (cAMP), protein kinase A (PKA), and cAMP response element-binding protein (CREB) signaling pathways. First, it was found that 50 Hz 0.6 millitesla (mT) PEMFs promoted osteogenic differentiation of rat calvarial osteoblasts (ROBs), and that PEMFs activated cAMP-PKA-CREB signaling by increasing intracellular cAMP levels, facilitating phosphorylation of PKA and CREB, and inducing nuclear translocation of phosphorylated (p)-CREB. Blocking the signaling by adenylate cyclase (AC) and PKA inhibitors both abolished the osteogenic effect of PEMFs. Second, expression of sAC isoform was found to be increased significantly by PEMF treatment. Blocking sAC using sAC-specific inhibitor KH7 dramatically inhibited the osteogenic differentiation of ROBs. Finally, the peak bone mass of growing rats was significantly increased after 2 months of PEMF treatment with 90 min/day. The serum cAMP content, p-PKA, and p-CREB as well as the sAC protein expression levels were all increased significantly in femurs of treated rats. The current study indicated that PEMFs promote bone formation in vitro and in vivo by activating sAC-cAMP-PKA-CREB signaling pathway of osteoblasts directly or indirectly.
Collapse
Affiliation(s)
- Yuan-Yuan Wang
- Department of Bioengineering, School of Life Science and Engineering, Lanzhou University of Technology, Lanzhou, China
- Institute of Orthopaedics, Lanzhou General Hospital of CPLA, Lanzhou, China
| | - Xiu-Ying Pu
- Department of Bioengineering, School of Life Science and Engineering, Lanzhou University of Technology, Lanzhou, China
| | - Wen-Gui Shi
- Institute of Orthopaedics, Lanzhou General Hospital of CPLA, Lanzhou, China
| | - Qing-Qing Fang
- Institute of Orthopaedics, Lanzhou General Hospital of CPLA, Lanzhou, China
| | - Xin-Ru Chen
- Department of Biology, College of Life Sciences, Northwest A & F University, Yanglin, China
| | - Hui-Rong Xi
- Institute of Orthopaedics, Lanzhou General Hospital of CPLA, Lanzhou, China
| | - Yu-Hai Gao
- Institute of Orthopaedics, Lanzhou General Hospital of CPLA, Lanzhou, China
| | - Jian Zhou
- Institute of Orthopaedics, Lanzhou General Hospital of CPLA, Lanzhou, China
| | - Cory J Xian
- Sansom Institute for Health Research, School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, South Australia, Australia
| | - Ke-Ming Chen
- Institute of Orthopaedics, Lanzhou General Hospital of CPLA, Lanzhou, China
| |
Collapse
|
42
|
|
43
|
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: 5.2] [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
|
44
|
Azadian E, Arjmand B, Khodaii Z, Ardeshirylajimi A. A comprehensive overview on utilizing electromagnetic fields in bone regenerative medicine. Electromagn Biol Med 2019; 38:1-20. [PMID: 30661411 DOI: 10.1080/15368378.2019.1567527] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Stem cells are one of the most important sources to develope a new strategy for repairing bone lesions through tissue engineering. Osteogenic differentiation of stem cells can be affected by various factors such as biological, chemical, physiological, and physical ones. The application of ELF-EMFs has been the subject of many research in bone tissue engineering and evidence suggests that this exogenous physical stimulus can promote osteogenic differentiation in several types of cells. The purpose of this paper is to review the current knowledge on the effects of EMFs on stem cells in bone tissue engineering studies. We recapitulated and analyzed 39 articles that were focused on the application of EMFs for bone tissue engineering purposes. We tabulated scattered information from these articles for easy use and tried to provide an overview of conducted research and identify the knowledge gaps in the field.
Collapse
Affiliation(s)
- Esmaeel Azadian
- a Urogenital Stem Cell Research Center , Shahid Beheshti University of Medical Sciences , Tehran , Iran.,b Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine , Shahid Beheshti University of Medical Sciences , Tehran , Iran
| | - Bahar Arjmand
- a Urogenital Stem Cell Research Center , Shahid Beheshti University of Medical Sciences , Tehran , Iran.,b Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine , Shahid Beheshti University of Medical Sciences , Tehran , Iran
| | - Zohreh Khodaii
- c Dietary supplements and Probiotics research center , Alborz University of Medical Sciences , Karaj , Iran.,d Department of Biochemistry, Genetics and Nutrition, Faculty of Medicine , Alborz University of Medical Sciences , Karaj , Iran
| | - Abdolreza Ardeshirylajimi
- a Urogenital Stem Cell Research Center , Shahid Beheshti University of Medical Sciences , Tehran , Iran.,b Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine , Shahid Beheshti University of Medical Sciences , Tehran , Iran
| |
Collapse
|
45
|
Ort C, Dayekh K, Xing M, Mequanint K. Emerging Strategies for Stem Cell Lineage Commitment in Tissue Engineering and Regenerative Medicine. ACS Biomater Sci Eng 2018; 4:3644-3657. [PMID: 33429592 DOI: 10.1021/acsbiomaterials.8b00532] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Stem cells have transformed the fields of tissue engineering and regenerative medicine, and their potential to further advance these fields cannot be overstated. The stem cell niche is a dynamic microenvironment that determines cell fate during development and tissue repair following an injury. Classically, stem cells were studied in isolation of their microenvironment; however, contemporary research has produced a myriad of evidence that shows the importance of multiple aspects of the stem cell niche in regulating their processes. In the context of tissue engineering and regenerative medicine studies, the niche is an artificial environment provided by culture conditions. In vitro culture conditions may involve coculturing with other cell types, developing specific biomaterials, and applying relevant forces to promote the desired lineage commitment. Considerable advance has been made over the past few years toward directed stem cell differentiation; however, the unspecific differentiation of stem cells yielding a mixed population of cells has been a challenge. In this review, we provide a systematic review of the emerging strategies used for lineage commitment within the context of tissue engineering and regenerative medicine. These strategies include scaffold pore-size and pore-shape gradients, stress relaxation, sonic and electromagnetic effects, and magnetic forces. Finally, we provide insights and perspectives into future directions focusing on signaling pathways activated during lineage commitment using external stimuli.
Collapse
Affiliation(s)
| | | | - Malcolm Xing
- Department of Mechanical Engineering, University of Manitoba, 66 Chancellors Circle, Winnipeg R3T 2N2, Canada
| | | |
Collapse
|
46
|
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: 21] [Impact Index Per Article: 3.5] [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
|
47
|
Ferroni L, Gardin C, Dolkart O, Salai M, Barak S, Piattelli A, Amir-Barak H, Zavan B. Pulsed electromagnetic fields increase osteogenetic commitment of MSCs via the mTOR pathway in TNF-α mediated inflammatory conditions: an in-vitro study. Sci Rep 2018; 8:5108. [PMID: 29572540 PMCID: PMC5865106 DOI: 10.1038/s41598-018-23499-9] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Accepted: 03/14/2018] [Indexed: 12/31/2022] Open
Abstract
Pulsed electromagnetic fields (PEMFs) have been considered a potential treatment modality for fracture healing, however, the mechanism of their action remains unclear. Mammalian target of rapamycin (mTOR) signaling may affect osteoblast proliferation and differentiation. This study aimed to assess the osteogenic differentiation of mesenchymal stem cells (MSCs) under PEMF stimulation and the potential involvement of mTOR signaling pathway in this process. PEMFs were generated by a novel miniaturized electromagnetic device. Potential changes in the expression of mTOR pathway components, including receptors, ligands and nuclear target genes, and their correlation with osteogenic markers and transcription factors were analyzed. Involvement of the mTOR pathway in osteogenesis was also studied in the presence of proinflammatory mediators. PEMF exposure increased cell proliferation and adhesion and the osteogenic commitment of MSCs even in inflammatory conditions. Osteogenic-related genes were over-expressed following PEMF treatment. Our results confirm that PEMFs contribute to activation of the mTOR pathway via upregulation of the proteins AKT, MAPP kinase, and RRAGA, suggesting that activation of the mTOR pathway is required for PEMF-stimulated osteogenic differentiation. Our findings provide insights into how PEMFs influence osteogenic differentiation in normal and inflammatory environments.
Collapse
Affiliation(s)
- Letizia Ferroni
- Department of Biomedical Sciences, University of Padova, Via G. Colombo 3, 35100, Padova, Italy
| | - Chiara Gardin
- Department of Biomedical Sciences, University of Padova, Via G. Colombo 3, 35100, Padova, Italy
| | - Oleg Dolkart
- Division of Orthopaedic Surgery, Tel Aviv Sourasky Medical Center, Tel Aviv University Sackler Faculty of Medicine, Tel Aviv, Israel.
| | - Moshe Salai
- Division of Orthopaedic Surgery, Tel Aviv Sourasky Medical Center, Tel Aviv University Sackler Faculty of Medicine, Tel Aviv, Israel
| | | | - Adriano Piattelli
- Department of Medical, Oral, and Biotechnological Sciences, University of Chieti-Pescara, Chieti, Italy
| | - Hadar Amir-Barak
- Department of Internal Medicine E, Tel Aviv Sourasky Medical Center, Tel Aviv University Sackler Faculty of Medicine, Tel Aviv, Israel
| | - Barbara Zavan
- Department of Biomedical Sciences, University of Padova, Via G. Colombo 3, 35100, Padova, Italy
| |
Collapse
|
48
|
The Application of Pulsed Electromagnetic Fields (PEMFs) for Bone Fracture Repair: Past and Perspective Findings. Ann Biomed Eng 2018; 46:525-542. [DOI: 10.1007/s10439-018-1982-1] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Accepted: 01/12/2018] [Indexed: 12/29/2022]
|
49
|
Physical stimulation and scaffold composition efficiently support osteogenic differentiation of mesenchymal stem cells. Tissue Cell 2017; 50:1-7. [PMID: 29429509 DOI: 10.1016/j.tice.2017.11.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Revised: 11/18/2017] [Accepted: 11/19/2017] [Indexed: 01/26/2023]
Abstract
BACKGROUND Despite significant achievements in the field of tissue engineering, simplification and improvement of the existing protocols are of great importance. The use of complex differentiation media, due to the presence of multiple factors, may have some undesired effects on cell health and functions. Thus, minimizing the number of involved factors, while maintaining the differentiation efficiency, provides less costly and controllable conditions. Adipose-derived Mesenchymal stem cells (ASCs), the adult stem cells present in adipose tissue, can be a suitable source of stem cells due to abundant and ease of access. The aim of this study is to optimize the osteogenic differentiation of ASCs by chemical composition of scaffold, in the first step, and then by electromagnetic treatments. METHODS ASCs were cultured on PVA/PES scaffold and tissue culture polystyrene surfaces (TCPS) and osteogenic differentiation was performed with either osteogenic medium, or electromagnetic field or both. The impact of each treatment on ASCs growth and proliferation was measured by MTT assay. Changes in gene expression levels of osteogenic-specific markers including ALP and RUNX2 were determined by Real Time PCR. Furthermore, alkaline phosphatase activity and calcium deposition were measured. RESULTS The MTT assay showed the significant effects on cell growth and respiration in scaffold-seeded ASCs treated with electromagnetic field, compared to control TCPS plate. Also, the electromagnetic treatment, increased alkaline phosphatase activity and calcium deposition. Finally, Real Time PCR showed higher expression of ALP and RUNX2 genes in electromagnetic field groups compared to control groups. CONCLUSION It can be concluded that PVA/PES scaffold used in this study improved the osteogenic capacity of ASCs. Moreover, the osteogenic potential of ASCs seeded on PVA/PES scaffold could be augmented by electromagnetic field without any chemical stimulation.
Collapse
|
50
|
Tschon M, Veronesi F, Contartese D, Sartori M, Martini L, Vincenzi F, Ravani A, Varani K, Fini M. Effects of pulsed electromagnetic fields and platelet rich plasma in preventing osteoclastogenesis in an in vitro model of osteolysis. J Cell Physiol 2017; 233:2645-2656. [PMID: 28786478 DOI: 10.1002/jcp.26143] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Accepted: 08/07/2017] [Indexed: 12/17/2022]
Abstract
Osteolysis is the main limiting cause for the survival of an orthopedic prosthesis and is accompanied by an enhancement in osteoclastogenesis and inflammation, due by wear debris formation. Unfortunately therapeutic treatments, besides revision surgery, are not available. The aim of the present study was to evaluate the effects of Pulsed Electro Magnetic Fields (PEMFs) and platelet rich plasma (PRP), alone or in combination, in an in vitro model of osteolysis. Rats peripheral blood mononuclear cells were cultured on Ultra High Molecular Weight Polyethylene particles and divided into four groups of treatments: (1) PEMF stimulation (12 hr/day, 2.5 mT, 75 Hz, 1.3 ms pulse duration); (2) 10% PRP; (3) combination of PEMFs, and PRP; (4) no treatment. Treatments were performed for 3 days and cell viability, osteoclast number, expression of genes related to osteoclastogenesis and inflammation and production of pro-inflammatory cytokines were assessed up to 14 days. PEMF stimulation exerted best results because it increased cell viability at early time points and counteracted osteoclastogenesis at 14 days. On the contrary, PRP increased osteoclastogenesis and reduced cell viability in comparison to PEMFs alone. The combination of PEMFs and PRP increased cell viability over time and reduced osteoclastogenesis in comparison to PRP alone. However, these positive results did not exceed the level achieved by PEMF alone. At longer time points PEMF could not counteract osteoclastogenesis increased by PRP. Regarding inflammation, all treatments maintained the production of pro-inflammatory cytokines at low level, although PRP increased the level of interleukin 1 beta.
Collapse
Affiliation(s)
- Matilde Tschon
- Laboratory of Preclinical and Surgical Studies, Rizzoli Orthopedic Institute, Bologna, Italy
| | - Francesca Veronesi
- Laboratory of Preclinical and Surgical Studies, Rizzoli Orthopedic Institute, Bologna, Italy
| | - Deyanira Contartese
- Laboratory of Preclinical and Surgical Studies, Rizzoli Orthopedic Institute, Bologna, Italy
| | - Maria Sartori
- Laboratory of Biocompatibility, Technological Innovations and Advanced Therapies, Research Innovation and Technology Department (RIT), Rizzoli Orthopaedic Institute, Bologna, Italy
| | - Lucia Martini
- Laboratory of Preclinical and Surgical Studies, Rizzoli Orthopedic Institute, Bologna, Italy
| | - Fabrizio Vincenzi
- Department of Medical Sciences, Laboratory of Cellular and Molecular Pharmacology, University of Ferrara, Ferrara, Italy
| | - Annalisa Ravani
- Department of Medical Sciences, Laboratory of Cellular and Molecular Pharmacology, University of Ferrara, Ferrara, Italy
| | - Katia Varani
- Department of Medical Sciences, Laboratory of Cellular and Molecular Pharmacology, University of Ferrara, Ferrara, Italy
| | - Milena Fini
- Laboratory of Preclinical and Surgical Studies, Rizzoli Orthopedic Institute, Bologna, Italy
| |
Collapse
|