1
|
Amjad MD, Marramiero L, Romasco T, Cipollina A, Hossein HHS, Mantarro M, Piattelli A, Fulle S, Di Pietro N, Mancinelli R. Complex Magnetic Fields: Harnessing the Electromagnetic Symphony for Possible Applications in Regenerative Medicine and Antifungal Properties. Int Wound J 2025; 22:e70679. [PMID: 40390303 PMCID: PMC12089650 DOI: 10.1111/iwj.70679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 03/10/2025] [Accepted: 04/29/2025] [Indexed: 05/21/2025] Open
Abstract
Complex magnetic fields (CMFs) represent an emerging frontier in regenerative medicine, offering significant potential for innovative therapeutic strategies. This review examined both the theoretical foundations and practical applications of CMFs, focusing on their roles in tissue regeneration and antifungal activity. A comprehensive review of electronic databases (PubMed, Scopus, and Embase) identified seven pivotal studies on in vitro models concerning the CMF topic. Although the number of studies is limited, they collectively highlighted the promising therapeutic potential of CMFs in enhancing wound healing, reducing oxidative stress, and neuroinflammation in diabetic neuropathy, positively influencing mitochondrial function, modulating immune responses, promoting cellular communication, inhibiting the growth and adhesion of Candida albicans to medical surfaces, and enhancing dental pulp stem cell proliferation under inflammatory conditions. These findings suggested that CMFs may offer an eco-sustainable approach, effectively targeting pathogens while preserving human cell integrity. While the current body of research is insightful, it remains in its early stages. To fully leverage the therapeutic potential of CMFs, more comprehensive studies are needed to refine their application and confirm their effectiveness across diverse clinical scenarios. This is essential for integrating CMFs into clinical practice, where they promise to revolutionise treatment approaches.
Collapse
Affiliation(s)
- Muhammad Dawood Amjad
- Department of Medical, Oral and Biotechnological Sciences“G. D'Annunzio” University of Chieti‐PescaraChietiItaly
| | - Lorenzo Marramiero
- Department of Neurosciences, Imaging and Clinical Sciences“G. D'Annunzio” University of Chieti‐PescaraChietiItaly
| | - Tea Romasco
- Department of Medical, Oral and Biotechnological Sciences“G. D'Annunzio” University of Chieti‐PescaraChietiItaly
- Center for Advanced Studies and Technology‐CAST, “G. D'Annunzio” University of Chieti‐PescaraChietiItaly
| | | | - Hamid Heydari Sheikh Hossein
- Department of Medical, Oral and Biotechnological Sciences“G. D'Annunzio” University of Chieti‐PescaraChietiItaly
| | | | - Adriano Piattelli
- School of DentistrySaint Camillus International University of Health and Medical SciencesRomeItaly
- Facultad de MedicinaUniversidad Católica San Antonio de Murcia (UCAM)MurciaSpain
| | - Stefania Fulle
- Department of Neurosciences, Imaging and Clinical Sciences“G. D'Annunzio” University of Chieti‐PescaraChietiItaly
| | - Natalia Di Pietro
- Department of Medical, Oral and Biotechnological Sciences“G. D'Annunzio” University of Chieti‐PescaraChietiItaly
- Center for Advanced Studies and Technology‐CAST, “G. D'Annunzio” University of Chieti‐PescaraChietiItaly
| | - Rosa Mancinelli
- Department of Neurosciences, Imaging and Clinical Sciences“G. D'Annunzio” University of Chieti‐PescaraChietiItaly
| |
Collapse
|
2
|
Liu Y, Tang Q, Tao Q, Dong H, Shi Z, Zhou L. Low-frequency magnetic field therapy for glioblastoma: Current advances, mechanisms, challenges and future perspectives. J Adv Res 2025; 69:531-543. [PMID: 38565404 PMCID: PMC11954840 DOI: 10.1016/j.jare.2024.03.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Revised: 03/10/2024] [Accepted: 03/29/2024] [Indexed: 04/04/2024] Open
Abstract
BACKGROUND Glioblastoma (GBM) is the most common malignant tumour of the central nervous system. Despite recent advances in multimodal GBM therapy incorporating surgery, radiotherapy, systemic therapy (chemotherapy, targeted therapy), and supportive care, the overall survival (OS) remains poor, and long-term survival is rare. Currently, the primary obstacles hindering the effectiveness of GBM treatment are still the blood-brain barrier and tumor heterogeneity. In light of its substantial advantages over conventional therapies, such as strong penetrative ability and minimal side effects, low-frequency magnetic fields (LF-MFs) therapy has gradually caught the attention of scientists. AIM OF REVIEW In this review, we shed the light on the current status of applying LF-MFs in the treatment of GBM. We specifically emphasize our current understanding of the mechanisms by which LF-MFs mediate anticancer effects and the challenges faced by LF-MFs in treating GBM cells. Furthermore, we discuss the prospective applications of magnetic field therapy in the future treatment of GBM. Key scientific concepts of review: The review explores the current progress on the use of LF-MFs in the treatment of GBM with a special focus on the potential underlying mechanisms of LF-MFs in anticancer effects. Additionally, we also discussed the complex magnetic field features and biological characteristics related to magnetic bioeffects. Finally, we proposed a promising magnetic field treatment strategy for future applications in GBM therapy.
Collapse
Affiliation(s)
- Yinlong Liu
- Department of Neurosurgery, Huashan Hospital, Fudan University, China
| | - Qisheng Tang
- Department of Neurosurgery, Huashan Hospital, Fudan University, China; National Center for Neurological Disorders, China; Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, China; Neurosurgical Institute of Fudan University, Shanghai, China; Shanghai Clinical Medical Center of Neurosurgery, China
| | - Quan Tao
- Shanghai Institute of Microsystem and Information Technology, China
| | - Hui Dong
- Shanghai Institute of Microsystem and Information Technology, China
| | - Zhifeng Shi
- Department of Neurosurgery, Huashan Hospital, Fudan University, China; National Center for Neurological Disorders, China; Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, China; Neurosurgical Institute of Fudan University, Shanghai, China; Shanghai Clinical Medical Center of Neurosurgery, China.
| | - Liangfu Zhou
- Department of Neurosurgery, Huashan Hospital, Fudan University, China; National Center for Neurological Disorders, China; Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, China; Neurosurgical Institute of Fudan University, Shanghai, China; Shanghai Clinical Medical Center of Neurosurgery, China.
| |
Collapse
|
3
|
Djamgoz MBA. Stemness of Cancer: A Study of Triple-negative Breast Cancer From a Neuroscience Perspective. Stem Cell Rev Rep 2025; 21:337-350. [PMID: 39531198 PMCID: PMC11872763 DOI: 10.1007/s12015-024-10809-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/21/2024] [Indexed: 11/16/2024]
Abstract
Stemness, giving cancer cells massive plasticity enabling them to survive in dynamic (e.g. hypoxic) environments and become resistant to treatment, especially chemotherapy, is an important property of aggressive tumours. Here, we review some essentials of cancer stemness focusing on triple-negative breast cancer (TNBC), the most aggressive form of all breast cancers. TNBC cells express a range of genes and mechanisms associated with stemness, including the fundamental four "Yamanaka factors". Most of the evidence concerns the transcription factor / oncogene c-Myc and an interesting case is the expression of the neonatal splice variant of voltage-gated sodium channel subtype Nav1.5. On the whole, measures that reduce the stemness make cancer cells less aggressive, reducing their invasive/metastatic potential and increasing/restoring their chemosensitivity. Such measures include gene silencing techniques, epigenetic therapies as well as novel approaches like optogenetics aiming to modulate the plasma membrane voltage. Indeed, simply hyperpolarizing their membrane potential can make stem cells differentiate. Finally, we give an overview of the clinical aspects and exploitation of cancer/TNBC stemness, including diagnostics and therapeutics. In particular, personalised mRNA-based therapies and mechanistically meaningful combinations are promising and the emerging discipline of 'cancer neuroscience' is providing novel insights to both fundamental issues and clinical applications.
Collapse
Affiliation(s)
- Mustafa B A Djamgoz
- Department of Life Sciences, Imperial College London, South Kensington Campus, London, SW7 2AZ, UK.
| |
Collapse
|
4
|
Kaadan A, Salati S, Setti S, Aaron R. Augmentation of Deficient Bone Healing by Pulsed Electromagnetic Fields-From Mechanisms to Clinical Outcomes. Bioengineering (Basel) 2024; 11:1223. [PMID: 39768041 PMCID: PMC11672986 DOI: 10.3390/bioengineering11121223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 11/19/2024] [Accepted: 11/26/2024] [Indexed: 01/11/2025] Open
Abstract
Pulsed Electromagnetic Fields (PEMF) are widely used, with excellent clinical outcomes. However, their mechanism of action has not yet been completely understood. The purpose of this review is to describe current observations on the mechanisms of PEMF, together with its clinical efficacy. Osteoblast responsiveness to PEMF is described on several scales, from the cell membrane to clinically relevant bone formation. PEMF has been shown to activate membrane adenosine receptors. The role of adenosine receptors in activating intracellular second messenger pathways, such as the canonical Wnt/β-catenin pathway and the mitogen-activated protein kinases (MAPK) pathway, is described. The responsiveness of osteoblasts and the synthesis of structural and signaling proteins constitute the role of PEMFs in promoting osteogenesis and bone matrix synthesis, and they are described. Multiple studies, ranging from observational and randomized to meta-analyses that investigate the clinical efficacy of PEMF, are described. This review presents a favorable conclusion on the clinical effects of PEMF while unlocking the "black box" of PEMF's mechanism of action, thus improving confidence in the clinical utility of PEMF in bone repair.
Collapse
Affiliation(s)
- Amr Kaadan
- Department of Orthopedic Surgery, Warren Alpert Medical School of Brown University, Providence, RI 02903, USA;
| | - Simona Salati
- Medical Division, Igea S.p.A, 41012 Carpi, Italy; (S.S.); (S.S.)
| | - Stefania Setti
- Medical Division, Igea S.p.A, 41012 Carpi, Italy; (S.S.); (S.S.)
| | - Roy Aaron
- Department of Orthopedic Surgery, Warren Alpert Medical School of Brown University, Providence, RI 02903, USA;
| |
Collapse
|
5
|
Gerdesmeyer L, Tübel J, Obermeier A, Harrasser N, Glowalla C, von Eisenhart-Rothe R, Burgkart R. Extracorporeal Magnetotransduction Therapy as a New Form of Electromagnetic Wave Therapy: From Gene Upregulation to Accelerated Matrix Mineralization in Bone Healing. Biomedicines 2024; 12:2269. [PMID: 39457582 PMCID: PMC11505246 DOI: 10.3390/biomedicines12102269] [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: 09/11/2024] [Revised: 09/28/2024] [Accepted: 09/30/2024] [Indexed: 10/28/2024] Open
Abstract
BACKGROUND Electromagnetic field therapy is gaining attention for its potential in treating bone disorders, with Extracorporeal Magnetotransduction Therapy (EMTT) emerging as an innovative approach. EMTT offers a higher oscillation frequency and magnetic field strength compared to traditional Pulsed Electromagnetic Field (PEMF) therapy, showing promise in enhancing fracture healing and non-union recovery. However, the mechanisms underlying these effects remain unclear. RESULTS This study demonstrates that EMTT significantly enhances osteoblast bone formation at multiple levels, from gene expression to extracellular matrix mineralization. Key osteoblastogenesis regulators, including SP7 and RUNX2, and bone-related genes such as COL1A1, ALPL, and BGLAP, were upregulated, with expression levels surpassing those of the control group by over sevenfold (p < 0.001). Enhanced collagen synthesis and mineralization were confirmed by von Kossa and Alizarin Red staining, indicating increased calcium and phosphate deposition. Additionally, calcium imaging revealed heightened calcium influx, suggesting a cellular mechanism for EMTT's osteogenic effects. Importantly, EMTT did not compromise cell viability, as confirmed by live/dead staining and WST-1 assays. CONCLUSION This study is the first to show that EMTT can enhance all phases of osteoblastogenesis and improve the production of critical mineralization components, offering potential clinical applications in accelerating fracture healing, treating osteonecrosis, and enhancing implant osseointegration.
Collapse
Affiliation(s)
- Lennart Gerdesmeyer
- Department of Orthopaedics and Sports Orthopaedics, Klinikum rechts der Isar, Technical University of Munich, Ismaninger Str. 22, 81675 Munich, Germany
| | - Jutta Tübel
- Department of Orthopaedics and Sports Orthopaedics, Klinikum rechts der Isar, Technical University of Munich, Ismaninger Str. 22, 81675 Munich, Germany
| | - Andreas Obermeier
- Department of Orthopaedics and Sports Orthopaedics, Klinikum rechts der Isar, Technical University of Munich, Ismaninger Str. 22, 81675 Munich, Germany
| | - Norbert Harrasser
- Department of Orthopaedics and Sports Orthopaedics, Klinikum rechts der Isar, Technical University of Munich, Ismaninger Str. 22, 81675 Munich, Germany
- ECOM Excellent Center of Medicine, Arabellastraße 17, 81925 Munich, Germany
| | - Claudio Glowalla
- Department of Orthopaedics and Sports Orthopaedics, Klinikum rechts der Isar, Technical University of Munich, Ismaninger Str. 22, 81675 Munich, Germany
- BG Unfallklinik Murnau, Professor-Küntscher-Straße 8, 82418 Murnau am Staffelsee, Germany
| | - Rüdiger von Eisenhart-Rothe
- Department of Orthopaedics and Sports Orthopaedics, Klinikum rechts der Isar, Technical University of Munich, Ismaninger Str. 22, 81675 Munich, Germany
| | - Rainer Burgkart
- Department of Orthopaedics and Sports Orthopaedics, Klinikum rechts der Isar, Technical University of Munich, Ismaninger Str. 22, 81675 Munich, Germany
| |
Collapse
|
6
|
Martins LA, García-Parra N, Ródenas-Rochina J, Cordón L, Sempere A, Ribeiro C, Lanceros-Méndez S, Gómez-Ribelles JL. Assemblable 3D biomimetic microenvironment for hMSC osteogenic differentiation. Biomed Mater 2024; 19:065013. [PMID: 39303743 DOI: 10.1088/1748-605x/ad7dc4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Accepted: 09/20/2024] [Indexed: 09/22/2024]
Abstract
Adequate simulation mimicking a tissue's native environment is one of the elemental premises in tissue engineering. Although various attempts have been made to induce human mesenchymal stem cells (hMSC) into an osteogenic pathway, they are still far from widespread clinical application. Most strategies focus primarily on providing a specific type of cue, inadequately replicating the complexity of the bone microenvironment. An alternative multifunctional platform for hMSC osteogenic differentiation has been produced. It is based on poly(vinylidene fluoride) (PVDF) and cobalt ferrites magnetoelectric microspheres, functionalized with collagen and gelatin, and packed in a 3D arrangement. This platform is capable of performing mechanical stimulation of piezoelectric PVDF, mimicking the bones electromechanical biophysical cues. Surface functionalization with extracellular matrix biomolecules and osteogenic medium complete this all-round approach. hMSC were cultured in osteogenic inducing conditions and tested for proliferation, surface biomarkers, and gene expression to evaluate their osteogenic commitment.
Collapse
Affiliation(s)
- Luis A Martins
- Center for Biomaterials and Tissue Engineering, CBIT, Universitat Politècnica de València, 46022 Valencia, Spain
| | - Nadia García-Parra
- Center for Biomaterials and Tissue Engineering, CBIT, Universitat Politècnica de València, 46022 Valencia, Spain
| | - Joaquín Ródenas-Rochina
- Center for Biomaterials and Tissue Engineering, CBIT, Universitat Politècnica de València, 46022 Valencia, Spain
| | - Lourdes Cordón
- Hematology Research Group, Instituto de Investigación Sanitaria La Fe (IISLAFE), 46022 Valencia, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Instituto Carlos III, 28029 Madrid, Spain
| | - Amparo Sempere
- Hematology Research Group, Instituto de Investigación Sanitaria La Fe (IISLAFE), 46022 Valencia, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Instituto Carlos III, 28029 Madrid, Spain
- Hematology Department, Hospital Universitari i Politècnic La Fe, 46026 Valencia, Spain
| | - Clarisse Ribeiro
- CF-UM-UP-Physics Centre of Minho and Porto Universities, University of Minho, 4710-057 Braga, Portugal
- LaPMET-Laboratory of Physics for Materials and Emergent Technologies, University of Minho, 4710-057 Braga, Portugal
| | - Senentxu Lanceros-Méndez
- CF-UM-UP-Physics Centre of Minho and Porto Universities, University of Minho, 4710-057 Braga, Portugal
- BCMaterials-Basque Center for Materials Applications and Nanostructures, University of the Basque Country, 48940 Leioa, Spain
- IKERBASQUE, Basque Foundation for Science, 48013 Bilbao, Spain
| | - José Luis Gómez-Ribelles
- Center for Biomaterials and Tissue Engineering, CBIT, Universitat Politècnica de València, 46022 Valencia, Spain
- Centro de Investigación Biomédica en Red de Bioingeniería Biomateriales y Nanomedicina (CIBER-BBN), Instituto de Salud Carlos III, 28029 Madrid, Spain
| |
Collapse
|
7
|
Torretta E, Moriggi M, Capitanio D, Orfei CP, Raffo V, Setti S, Cadossi R, de Girolamo L, Gelfi C. Effects of Pulsed Electromagnetic Field Treatment on Skeletal Muscle Tissue Recovery in a Rat Model of Collagenase-Induced Tendinopathy: Results from a Proteome Analysis. Int J Mol Sci 2024; 25:8852. [PMID: 39201538 PMCID: PMC11354614 DOI: 10.3390/ijms25168852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Revised: 08/09/2024] [Accepted: 08/12/2024] [Indexed: 09/02/2024] Open
Abstract
Tendon disorders often result in decreased muscle function and atrophy. Pulsed Electromagnetic Fields (PEMFs) have shown potential in improving tendon fiber structure and muscle recovery. However, the molecular effects of PEMF therapy on skeletal muscle, beyond conventional metrics like MRI or markers of muscle decline, remain largely unexplored. This study investigates the metabolic and structural changes in PEMF-treated muscle tissue using proteomics in a rat model of Achilles tendinopathy induced by collagenase. Sprague Dawley rats were unilaterally induced for tendinopathy with type I collagenase injection and exposed to PEMFs for 8 h/day. Gastrocnemius extracts from untreated or PEMF-treated rats were analyzed with LC-MS/MS, and proteomics differential analysis was conducted through label-free quantitation. PEMF-treated animals exhibited decreased glycolysis and increased LDHB expression, enhancing NAD signaling and ATP production, which boosted respiratory chain activity and fatty acid beta-oxidation. Antioxidant protein levels increased, controlling ROS production. PEMF therapy restored PGC1alpha and YAP levels, decreased by tendinopathy. Additionally, myosins regulating slow-twitch fibers and proteins involved in fiber alignment and force transmission increased, supporting muscle recovery and contractile function. Our findings show that PEMF treatment modulates NAD signaling and oxidative phosphorylation, aiding muscle recovery through the upregulation of YAP and PGC1alpha and increasing slow myosin isoforms, thus speeding up physiological recovery.
Collapse
Affiliation(s)
- Enrica Torretta
- Laboratory of Proteomics and Lipidomics, IRCCS Orthopedic Institute Galeazzi, 20161 Milan, Italy;
| | - Manuela Moriggi
- Department of Biomedical Sciences for Health, University of Milan, 20090 Segrate, Italy; (M.M.); (D.C.)
| | - Daniele Capitanio
- Department of Biomedical Sciences for Health, University of Milan, 20090 Segrate, Italy; (M.M.); (D.C.)
| | - Carlotta Perucca Orfei
- Orthopaedic Biotechnology Laboratory, IRCCS Orthopedic Institute Galeazzi, 20161 Milan, Italy (V.R.); (L.d.G.)
| | - Vincenzo Raffo
- Orthopaedic Biotechnology Laboratory, IRCCS Orthopedic Institute Galeazzi, 20161 Milan, Italy (V.R.); (L.d.G.)
| | | | | | - Laura de Girolamo
- Orthopaedic Biotechnology Laboratory, IRCCS Orthopedic Institute Galeazzi, 20161 Milan, Italy (V.R.); (L.d.G.)
| | - Cecilia Gelfi
- Laboratory of Proteomics and Lipidomics, IRCCS Orthopedic Institute Galeazzi, 20161 Milan, Italy;
- Department of Biomedical Sciences for Health, University of Milan, 20090 Segrate, Italy; (M.M.); (D.C.)
| |
Collapse
|
8
|
Sun J, Xie W, Wu Y, Li Z, Li Y. Accelerated Bone Healing via Electrical Stimulation. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024:e2404190. [PMID: 39115981 DOI: 10.1002/advs.202404190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 07/01/2024] [Indexed: 08/10/2024]
Abstract
Piezoelectric effect produces an electrical signal when stress is applied to the bone. When the integrity of the bone is destroyed, the biopotential within the defect site is reduced and several physiological responses are initiated to facilitate healing. During the healing of the bone defect, the bioelectric potential returns to normal levels. Treatment of fractures that exceed innate regenerative capacity or exhibit delayed healing requires surgical intervention for bone reconstruction. For bone defects that cannot heal on their own, exogenous electric fields are used to assist in treatment. This paper reviews the effects of exogenous electrical stimulation on bone healing, including osteogenesis, angiogenesis, reduction in inflammation and effects on the peripheral nervous system. This paper also reviews novel electrical stimulation methods, such as small power supplies and nanogenerators, that have emerged in recent years. Finally, the challenges and future trends of using electrical stimulation therapy for accelerating bone healing are discussed.
Collapse
Affiliation(s)
- Jianfeng Sun
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Wenqing Xie
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Yuxiang Wu
- School of Kinesiology, Jianghan University, Wuhan, Hubei, 430056, China
| | - Zhou Li
- Beijing Institute of Nanoenergy and Nanosystems, Chinese Academy of Sciences, Beijing, 101400, China
| | - Yusheng Li
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| |
Collapse
|
9
|
Anjum S, Wang Y, Xin Y, Li X, Li T, Zhang H, Quan L, Li Y, Arya DK, Rajinikanth P, Ao Q. Bioinspired core-shell nanofiber drug-delivery system modulates osteogenic and osteoclast activity for bone tissue regeneration. Mater Today Bio 2024; 26:101088. [PMID: 38779556 PMCID: PMC11109009 DOI: 10.1016/j.mtbio.2024.101088] [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: 02/12/2024] [Revised: 05/05/2024] [Accepted: 05/08/2024] [Indexed: 05/25/2024] Open
Abstract
Osteogenic-osteoclast coupling processes play a crucial role in bone regeneration. Recently, strategies that focus on multi-functionalized implant surfaces to enhance the healing of bone defects through the synergistic regulation of osteogenesis and osteoclastogenesis is still a challenging task in the field of bone tissue engineering. The aim of this study was to create a dual-drug release-based core-shell nanofibers with the intent of achieving a time-controlled release to facilitate bone regeneration. We fabricated core-shell P/PCL nanofibers using coaxial electrospinning, where alendronate (ALN) was incorporated into the core layer and hydroxyapatite (HA) into shell. The surface of the nanofiber construct was further modified with mussel-derived polydopamine (PDA) to induce hydrophilicity and enhance cell interactions. Surface characterizations confirmed the successful synthesis of PDA@PHA/PCL-ALN nanofibers endowed with excellent mechanical strength (20.02 ± 0.13 MPa) and hydrophilicity (22.56°), as well as the sustained sequential release of ALN and Ca ions. In vitro experiments demonstrated that PDA-functionalized core-shell PHA/PCL-ALN scaffolds possessed excellent cytocompatibility, enhanced cell adhesion and proliferation, alkaline phosphatase activity and osteogenesis-related genes. In addition to osteogenesis, the engineered scaffolds also significantly reduced osteoclastogenesis, such as tartrate-resistant acid phosphatase activity and osteoclastogenesis-related gene expression. After 12-week of implantation, it was observed that PDA@PHA/PCL-ALN nanofiber scaffolds, in a rat cranial defect model, significantly promoted bone repair and regeneration. Microcomputed tomography, histological examination, and immunohistochemical analysis collectively demonstrated that the PDA-functionalized core-shell PHA/PCL-ALN scaffolds exhibited exceptional osteogenesis-inducing and osteoclastogenesis-inhibiting effects. Finally, it may be concluded from our results that the bio-inspired surface-functionalized multifunctional, biomimetic and controlled release core-shell nanofiber provides a promising strategy to facilitate bone healing.
Collapse
Affiliation(s)
- Shabnam Anjum
- Department of Tissue Engineering, School of Intelligent Medicine, China Medical University, Shenyang, Liaoning, 110122, China
- NMPA Key Laboratory for Quality Research and Control of Tissue Regenerative Biomaterial & Institute of Regulatory Science for Medical Device & National Engineering Research Centre for Biomaterials, Sichuan University, Chengdu, Sichuan, 610064, China
| | - Yulin Wang
- NMPA Key Laboratory for Quality Research and Control of Tissue Regenerative Biomaterial & Institute of Regulatory Science for Medical Device & National Engineering Research Centre for Biomaterials, Sichuan University, Chengdu, Sichuan, 610064, China
| | - Yuan Xin
- NMPA Key Laboratory for Quality Research and Control of Tissue Regenerative Biomaterial & Institute of Regulatory Science for Medical Device & National Engineering Research Centre for Biomaterials, Sichuan University, Chengdu, Sichuan, 610064, China
| | - Xiao Li
- Department of Tissue Engineering, School of Intelligent Medicine, China Medical University, Shenyang, Liaoning, 110122, China
| | - Ting Li
- Department of Laboratory Medicine, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Hengtong Zhang
- NMPA Key Laboratory for Quality Research and Control of Tissue Regenerative Biomaterial & Institute of Regulatory Science for Medical Device & National Engineering Research Centre for Biomaterials, Sichuan University, Chengdu, Sichuan, 610064, China
| | - Liang Quan
- NMPA Key Laboratory for Quality Research and Control of Tissue Regenerative Biomaterial & Institute of Regulatory Science for Medical Device & National Engineering Research Centre for Biomaterials, Sichuan University, Chengdu, Sichuan, 610064, China
| | - Ya Li
- NMPA Key Laboratory for Quality Research and Control of Tissue Regenerative Biomaterial & Institute of Regulatory Science for Medical Device & National Engineering Research Centre for Biomaterials, Sichuan University, Chengdu, Sichuan, 610064, China
| | - Dilip Kumar Arya
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raebareli Road, Lucknow, India
| | - P.S. Rajinikanth
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raebareli Road, Lucknow, India
| | - Qiang Ao
- Department of Tissue Engineering, School of Intelligent Medicine, China Medical University, Shenyang, Liaoning, 110122, China
- NMPA Key Laboratory for Quality Research and Control of Tissue Regenerative Biomaterial & Institute of Regulatory Science for Medical Device & National Engineering Research Centre for Biomaterials, Sichuan University, Chengdu, Sichuan, 610064, China
| |
Collapse
|
10
|
Cianni L, Di Gialleonardo E, Coppola D, Capece G, Libutti E, Nannerini M, Maccauro G, Vitiello R. Current Evidence Using Pulsed Electromagnetic Fields in Osteoarthritis: A Systematic Review. J Clin Med 2024; 13:1959. [PMID: 38610722 PMCID: PMC11012419 DOI: 10.3390/jcm13071959] [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: 02/25/2024] [Revised: 03/22/2024] [Accepted: 03/25/2024] [Indexed: 04/14/2024] Open
Abstract
(1) Background: Osteoarthritis (OA) significantly impacts patients' quality of life and negatively affects public healthcare costs. The aim of this systematic review is to identify the effectiveness of pulsed electromagnetic fields (PEMFs) in OA treatment across different anatomical districts, determining pain reduction and overall improvement in the patient's quality of life. (2) Methods: In this systematic review following PRISMA guidelines, PubMed and Google Scholar were searched for randomized controlled trials involving patients with osteoarthritis undergoing PEMF therapy. Seventeen studies (1197 patients) were included. (3) Results: PEMF therapy demonstrated positive outcomes across various anatomical districts, primarily in knee osteoarthritis. Pain reduction, assessed through VAS and WOMAC scores, showed significant improvement (60% decrease in VAS, 42% improvement in WOMAC). The treatment duration varied (15 to 90 days), with diverse PEMF devices used. Secondary outcomes included improvements in quality of life, reduced medication usage, and enhanced physical function. (4) Conclusions: Diverse PEMF applications revealed promising results, emphasizing pain reduction and improvement in the quality of life of patients. The variability in the treatment duration and device types calls for further investigation. This review informs future research directions and potential advancements in optimizing PEMF therapies for diverse osteoarthritic manifestations.
Collapse
Affiliation(s)
- Luigi Cianni
- Orthopaedics and Trauma Surgery Unit, Catholic University of the Sacred Heart, 00168 Rome, Italy; (L.C.); (E.D.G.); (D.C.); (G.M.); (R.V.)
- Department of Ageing, Neurosciences, Head-Neck and Orthopedics Sciences, Orthopedics and Trauma Surgery Unit, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
| | - Emidio Di Gialleonardo
- Orthopaedics and Trauma Surgery Unit, Catholic University of the Sacred Heart, 00168 Rome, Italy; (L.C.); (E.D.G.); (D.C.); (G.M.); (R.V.)
| | - Donato Coppola
- Orthopaedics and Trauma Surgery Unit, Catholic University of the Sacred Heart, 00168 Rome, Italy; (L.C.); (E.D.G.); (D.C.); (G.M.); (R.V.)
| | - Giacomo Capece
- Orthopaedics and Trauma Surgery Unit, Catholic University of the Sacred Heart, 00168 Rome, Italy; (L.C.); (E.D.G.); (D.C.); (G.M.); (R.V.)
| | | | | | - Giulio Maccauro
- Orthopaedics and Trauma Surgery Unit, Catholic University of the Sacred Heart, 00168 Rome, Italy; (L.C.); (E.D.G.); (D.C.); (G.M.); (R.V.)
- Department of Ageing, Neurosciences, Head-Neck and Orthopedics Sciences, Orthopedics and Trauma Surgery Unit, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
| | - Raffaele Vitiello
- Orthopaedics and Trauma Surgery Unit, Catholic University of the Sacred Heart, 00168 Rome, Italy; (L.C.); (E.D.G.); (D.C.); (G.M.); (R.V.)
- Department of Ageing, Neurosciences, Head-Neck and Orthopedics Sciences, Orthopedics and Trauma Surgery Unit, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
| |
Collapse
|
11
|
Su DB, Zhao ZX, Yin DC, Ye YJ. Promising application of pulsed electromagnetic fields on tissue repair and regeneration. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2024; 187:36-50. [PMID: 38280492 DOI: 10.1016/j.pbiomolbio.2024.01.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 12/14/2023] [Accepted: 01/19/2024] [Indexed: 01/29/2024]
Abstract
Tissue repair and regeneration is a vital biological process in organisms, which is influenced by various internal mechanisms and microenvironments. Pulsed electromagnetic fields (PEMFs) are becoming a potential medical technology due to its advantages of effectiveness and non-invasiveness. Numerous studies have demonstrated that PEMFs can stimulate stem cell proliferation and differentiation, regulate inflammatory reactions, accelerate wound healing, which is of great significance for tissue regeneration and repair, providing a solid basis for enlarging its clinical application. However, some important issues such as optimal parameter system and potential deep mechanisms remain to be resolved due to PEMFs window effect and biological complexity. Thus, it is of great importance to comprehensively summarizing and analyzing the literature related to the biological effects of PEMFs in tissue regeneration and repair. This review expounded the biological effects of PEMFs on stem cells, inflammation response, wound healing and musculoskeletal disorders in order to improve the application value of PEMFs in medicine. It is believed that with the continuous exploration of biological effects of PEMFs, it will be applied increasingly widely to tissue repair and other diseases.
Collapse
Affiliation(s)
- Dan-Bo Su
- Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, 710072, China
| | - Zi-Xu Zhao
- Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, 710072, China
| | - Da-Chuan Yin
- Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, 710072, China
| | - Ya-Jing Ye
- Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, 710072, China.
| |
Collapse
|
12
|
Santos LF, Silva AS, Mano JF. Magnetic-Based Strategies for Regenerative Medicine and Tissue Engineering. Adv Healthc Mater 2023; 12:e2300605. [PMID: 37543723 DOI: 10.1002/adhm.202300605] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 08/03/2023] [Indexed: 08/07/2023]
Abstract
The fabrication of biological substitutes to repair, replace, or enhance tissue- and organ-level functions is a long-sought goal of tissue engineering (TE). However, the clinical translation of TE is hindered by several challenges, including the lack of suitable mechanical, chemical, and biological properties in one biomaterial, and the inability to generate large, vascularized tissues with a complex structure of native tissues. Over the past decade, a new generation of "smart" materials has revolutionized the conventional medical field, transforming TE into a more accurate and sophisticated concept. At the vanguard of scientific development, magnetic nanoparticles (MNPs) have garnered extensive attention owing to their significant potential in various biomedical applications owing to their inherent properties such as biocompatibility and rapid remote response to magnetic fields. Therefore, to develop functional tissue replacements, magnetic force-based TE (Mag-TE) has emerged as an alternative to conventional TE strategies, allowing for the fabrication and real-time monitoring of tissues engineered in vitro. This review addresses the recent studies on the use of MNPs for TE, emphasizing the in vitro, in vivo, and clinical applications. Future perspectives of Mag-TE in the fields of TE and regenerative medicine are also discussed.
Collapse
Affiliation(s)
- Lúcia F Santos
- Department of Chemistry, CICECO-Aveiro Institute of Materials, University of Aveiro, Aveiro, 3810-193, Portugal
| | - Ana S Silva
- Department of Chemistry, CICECO-Aveiro Institute of Materials, University of Aveiro, Aveiro, 3810-193, Portugal
| | - João F Mano
- Department of Chemistry, CICECO-Aveiro Institute of Materials, University of Aveiro, Aveiro, 3810-193, Portugal
| |
Collapse
|
13
|
Linnemann C, Sahin F, Chen Y, Falldorf K, Ronniger M, Histing T, Nussler AK, Ehnert S. NET Formation Was Reduced via Exposure to Extremely Low-Frequency Pulsed Electromagnetic Fields. Int J Mol Sci 2023; 24:14629. [PMID: 37834077 PMCID: PMC10572227 DOI: 10.3390/ijms241914629] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 09/20/2023] [Accepted: 09/22/2023] [Indexed: 10/15/2023] Open
Abstract
Fracture-healing is a highly complex and timely orchestrated process. Non-healing fractures are still a major clinical problem and treatment remains difficult. A 16 Hz extremely low-frequency pulsed electromagnetic field (ELF-PEMF) was identified as non-invasive adjunct therapy supporting bone-healing by inducing reactive oxygen species (ROS) and Ca2+-influx. However, ROS and Ca2+-influx may stimulate neutrophils, the first cells arriving at the wounded site, to excessively form neutrophil extracellular traps (NETs), which negatively affects the healing process. Thus, this study aimed to evaluate the effect of this 16 Hz ELF-PEMF on NET formation. Neutrophils were isolated from healthy volunteers and exposed to different NET-stimuli and the 16 Hz ELF-PEMF. NETs were quantified using Sytox Green Assay and immunofluorescence, Ca2+-influx and ROS with fluorescence probes. In contrast to mesenchymal cells, ELF-PEMF exposure did not induce ROS and Ca2+-influx in neutrophils. ELF-PEMF exposure did not result in basal or enhanced PMA-induced NET formation but did reduce the amount of DNA released. Similarly, NET formation induced by LPS and H2O2 was reduced through exposure to ELF-PEMF. As ELF-PEMF exposure did not induce NET release or negatively affect neutrophils, the ELF-PEMF exposure can be started immediately after fracture treatment.
Collapse
Affiliation(s)
- Caren Linnemann
- Siegfried Weller Institute for Trauma Research, BG Unfallklinik Tübingen, Eberhard Karls Universität Tuebingen, Schnarrenbergstraße 95, 72076 Tuebingen, Germany; (C.L.); (A.K.N.)
| | - Filiz Sahin
- Siegfried Weller Institute for Trauma Research, BG Unfallklinik Tübingen, Eberhard Karls Universität Tuebingen, Schnarrenbergstraße 95, 72076 Tuebingen, Germany; (C.L.); (A.K.N.)
| | - Yangmengfan Chen
- Siegfried Weller Institute for Trauma Research, BG Unfallklinik Tübingen, Eberhard Karls Universität Tuebingen, Schnarrenbergstraße 95, 72076 Tuebingen, Germany; (C.L.); (A.K.N.)
| | - Karsten Falldorf
- Sachtleben GmbH, Haus Spectrum am UKE, Martinistraße 64, 20251 Hamburg, Germany
| | - Michael Ronniger
- Sachtleben GmbH, Haus Spectrum am UKE, Martinistraße 64, 20251 Hamburg, Germany
| | - Tina Histing
- Siegfried Weller Institute for Trauma Research, BG Unfallklinik Tübingen, Eberhard Karls Universität Tuebingen, Schnarrenbergstraße 95, 72076 Tuebingen, Germany; (C.L.); (A.K.N.)
| | - Andreas K. Nussler
- Siegfried Weller Institute for Trauma Research, BG Unfallklinik Tübingen, Eberhard Karls Universität Tuebingen, Schnarrenbergstraße 95, 72076 Tuebingen, Germany; (C.L.); (A.K.N.)
| | - Sabrina Ehnert
- Siegfried Weller Institute for Trauma Research, BG Unfallklinik Tübingen, Eberhard Karls Universität Tuebingen, Schnarrenbergstraße 95, 72076 Tuebingen, Germany; (C.L.); (A.K.N.)
| |
Collapse
|
14
|
Han J, Ma Q, An Y, Wu F, Zhao Y, Wu G, Wang J. The current status of stimuli-responsive nanotechnologies on orthopedic titanium implant surfaces. J Nanobiotechnology 2023; 21:277. [PMID: 37596638 PMCID: PMC10439657 DOI: 10.1186/s12951-023-02017-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Accepted: 07/21/2023] [Indexed: 08/20/2023] Open
Abstract
With the continuous innovation and breakthrough of nanomedical technology, stimuli-responsive nanotechnology has been gradually applied to the surface modification of titanium implants to achieve brilliant antibacterial activity and promoted osteogenesis. Regarding to the different physiological and pathological microenvironment around implants before and after surgery, these surface nanomodifications are designed to respond to different stimuli and environmental changes in a timely, efficient, and specific way/manner. Here, we focus on the materials related to stimuli-responsive nanotechnology on titanium implant surface modification, including metals and their compounds, polymer materials and other materials. In addition, the mechanism of different response types is introduced according to different activation stimuli, including magnetic, electrical, photic, radio frequency and ultrasonic stimuli, pH and enzymatic stimuli (the internal stimuli). Meanwhile, the associated functions, potential applications and developing prospect were discussion.
Collapse
Affiliation(s)
- Jingyuan Han
- Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Engineering Research Center for Dental Materials and Advanced Manufacture, Department of Oral Implants, School of Stomatology, The Fourth Military Medical University, Xi’an, 710032 China
- School of Stomatology, Heilongjiang Key Lab of Oral Biomedicine Materials and Clinical Application, Experimental Center for Stomatology Engineering, Jiamusi University, Jiamusi, 154007 China
| | - Qianli Ma
- Department of Biomaterials, Institute of Clinical Dentistry, University of Oslo, Geitmyrsveien, Oslo, 710455 Norway
| | - Yanxin An
- Department of General Surgery, The First Affiliated Hospital of Xi’an Medical University, Xi’an, China
| | - Fan Wu
- Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Engineering Research Center for Dental Materials and Advanced Manufacture, Department of Oral Implants, School of Stomatology, The Fourth Military Medical University, Xi’an, 710032 China
- School of Stomatology, Heilongjiang Key Lab of Oral Biomedicine Materials and Clinical Application, Experimental Center for Stomatology Engineering, Jiamusi University, Jiamusi, 154007 China
| | - Yuqing Zhao
- Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Engineering Research Center for Dental Materials and Advanced Manufacture, Department of Oral Implants, School of Stomatology, The Fourth Military Medical University, Xi’an, 710032 China
- School of Stomatology, Heilongjiang Key Lab of Oral Biomedicine Materials and Clinical Application, Experimental Center for Stomatology Engineering, Jiamusi University, Jiamusi, 154007 China
| | - Gaoyi Wu
- School of Stomatology, Heilongjiang Key Lab of Oral Biomedicine Materials and Clinical Application, Experimental Center for Stomatology Engineering, Jiamusi University, Jiamusi, 154007 China
| | - Jing Wang
- Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Engineering Research Center for Dental Materials and Advanced Manufacture, Department of Oral Implants, School of Stomatology, The Fourth Military Medical University, Xi’an, 710032 China
| |
Collapse
|
15
|
Ma T, Ding Q, Liu C, Wu H. Electromagnetic fields regulate calcium-mediated cell fate of stem cells: osteogenesis, chondrogenesis and apoptosis. Stem Cell Res Ther 2023; 14:133. [PMID: 37194107 DOI: 10.1186/s13287-023-03303-w] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 03/28/2023] [Indexed: 05/18/2023] Open
Abstract
Electromagnetic fields (EMF) are increasing in popularity as a safe and non-invasive therapy. On the one hand, it is widely acknowledged that EMF can regulate the proliferation and differentiation of stem cells, promoting the undifferentiated cells capable of osteogenesis, angiogenesis, and chondroblast differentiation to achieve bone repair purpose. On the other hand, EMF can inhibit tumor stem cells proliferation and promote apoptosis to suppress tumor growth. As an essential second messenger, intracellular calcium plays a role in regulating cell cycle, such as proliferation, differentiation and apoptosis. There is increasing evidence that the modulation of intracellular calcium ion by EMF leads to differential outcomes in different stem cells. This review summarizes the regulation of channels, transporters, and ion pumps by EMF-induced calcium oscillations. It furtherly discusses the role of molecules and pathways activated by EMF-dependent calcium oscillations in promoting bone and cartilage repair and inhibiting tumor stem cells growth.
Collapse
Affiliation(s)
- Tian Ma
- Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Qing Ding
- Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Chaoxu Liu
- Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China.
| | - Hua Wu
- Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China.
| |
Collapse
|
16
|
Mazzotti A, Langone L, Artioli E, Zielli SO, Arceri A, Setti S, Leigheb M, Samaila EM, Faldini C. Applications and Future Perspective of Pulsed Electromagnetic Fields in Foot and Ankle Sport-Related Injuries. APPLIED SCIENCES 2023; 13:5807. [DOI: 10.3390/app13095807] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/23/2025]
Abstract
Foot and ankle injuries are common in many sports. One of the main athletes issues is the time for sport resumption after trauma. Recently, extensive efforts have been made to speed up the athletes’ return-to-sport and to prevent joint degeneration. Among the conservative treatment options, biophysical stimulation with pulsed electromagnetic fields (PEMFs) is listed. This narrative review aims to outline current applications of PEMFs in main foot and ankle sport-related injuries, in particular in the treatment of bone marrow edema, osteochondral defects, fractures, and nonunions. Despite further high-quality studies on foot and ankle injuries are needed, PEMFs seem to be a valid aid to enhance the endogenous osteogenesis, to resolve the bone marrow edema, to inhibit the joint inflammation, preserving articular cartilage degeneration, and to relieve pain.
Collapse
Affiliation(s)
- Antonio Mazzotti
- IRCCS Istituto Ortopedico Rizzoli, 1st Orthopaedics and Traumatology Clinic, University of Bologna, 40136 Bologna, Italy
- Department of Biomedical and Neuromotor Sciences, University of Bologna, 40126 Bologna, Italy
| | - Laura Langone
- IRCCS Istituto Ortopedico Rizzoli, 1st Orthopaedics and Traumatology Clinic, University of Bologna, 40136 Bologna, Italy
- Department of Biomedical and Neuromotor Sciences, University of Bologna, 40126 Bologna, Italy
| | - Elena Artioli
- IRCCS Istituto Ortopedico Rizzoli, 1st Orthopaedics and Traumatology Clinic, University of Bologna, 40136 Bologna, Italy
- Department of Biomedical and Neuromotor Sciences, University of Bologna, 40126 Bologna, Italy
| | - Simone Ottavio Zielli
- IRCCS Istituto Ortopedico Rizzoli, 1st Orthopaedics and Traumatology Clinic, University of Bologna, 40136 Bologna, Italy
- Department of Biomedical and Neuromotor Sciences, University of Bologna, 40126 Bologna, Italy
| | - Alberto Arceri
- IRCCS Istituto Ortopedico Rizzoli, 1st Orthopaedics and Traumatology Clinic, University of Bologna, 40136 Bologna, Italy
- Department of Biomedical and Neuromotor Sciences, University of Bologna, 40126 Bologna, Italy
| | | | - Massimiliano Leigheb
- Orthopaedics and Traumatology Unit, “Maggiore della Carità” Hospital, Department of Health Sciences, University of Piemonte Orientale, 28100 Novara, Italy
| | - Elena Manuela Samaila
- Department of Orthopedics and Trauma Surgery, University of Verona, Surgical Center “P. Confortini”, P.le A. Stefani, 1, 37126 Verona, Italy
| | - Cesare Faldini
- IRCCS Istituto Ortopedico Rizzoli, 1st Orthopaedics and Traumatology Clinic, University of Bologna, 40136 Bologna, Italy
- Department of Biomedical and Neuromotor Sciences, University of Bologna, 40126 Bologna, Italy
| |
Collapse
|
17
|
Czwartos J, Dobosz B, Kasprzycka W, Osuchowska PN, Stępińska M, Trafny EA, Starzyński J, Mierczyk Z. Preliminary Study on the Effect of a Single High-Energy Electromagnetic Pulse on Morphology and Free Radical Generation in Human Mesenchymal Stem Cells. Int J Mol Sci 2023; 24:ijms24087246. [PMID: 37108409 PMCID: PMC10139018 DOI: 10.3390/ijms24087246] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 03/28/2023] [Accepted: 04/11/2023] [Indexed: 04/29/2023] Open
Abstract
The effect of nanosecond electromagnetic pulses on human health, and especially on forming free radicals in human cells, is the subject of continuous research and ongoing discussion. This work presents a preliminary study on the effect of a single high-energy electromagnetic pulse on morphology, viability, and free radical generation in human mesenchymal stem cells (hMSC). The cells were exposed to a single electromagnetic pulse with an electric field magnitude of ~1 MV/m and a pulse duration of ~120 ns generated from a 600 kV Marx generator. The cell viability and morphology at 2 h and 24 h after exposure were examined using confocal fluorescent microscopy and scanning electron microscopy (SEM), respectively. The number of free radicals was investigated with electron paramagnetic resonance (EPR). The microscopic observations and EPR measurements showed that the exposure to the high-energy electromagnetic pulse influenced neither the number of free radicals generated nor the morphology of hMSC in vitro compared to control samples.
Collapse
Affiliation(s)
- Joanna Czwartos
- Institute of Optoelectronics, Military University of Technology, 2 Kaliskiego St., 00-908 Warsaw, Poland
| | - Bernadeta Dobosz
- Faculty of Physics, Adam Mickiewicz University, Uniwersytetu Poznańskiego 2, 61-614 Poznań, Poland
| | - Wiktoria Kasprzycka
- Institute of Optoelectronics, Military University of Technology, 2 Kaliskiego St., 00-908 Warsaw, Poland
| | - Paulina Natalia Osuchowska
- Institute of Optoelectronics, Military University of Technology, 2 Kaliskiego St., 00-908 Warsaw, Poland
| | - Małgorzata Stępińska
- Institute of Optoelectronics, Military University of Technology, 2 Kaliskiego St., 00-908 Warsaw, Poland
| | - Elżbieta Anna Trafny
- Institute of Optoelectronics, Military University of Technology, 2 Kaliskiego St., 00-908 Warsaw, Poland
| | - Jacek Starzyński
- Faculty of Electronical Engineering, Warsaw University of Technology, Koszykowa 75, 00-662 Warsaw, Poland
| | - Zygmunt Mierczyk
- Institute of Optoelectronics, Military University of Technology, 2 Kaliskiego St., 00-908 Warsaw, Poland
| |
Collapse
|
18
|
Chen Y, Braun BJ, Menger MM, Ronniger M, Falldorf K, Histing T, Nussler AK, Ehnert S. Intermittent Exposure to a 16 Hz Extremely Low Frequency Pulsed Electromagnetic Field Promotes Osteogenesis In Vitro through Activating Piezo 1-Induced Ca 2+ Influx in Osteoprogenitor Cells. J Funct Biomater 2023; 14:165. [PMID: 36976089 PMCID: PMC10055851 DOI: 10.3390/jfb14030165] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 03/14/2023] [Accepted: 03/15/2023] [Indexed: 03/29/2023] Open
Abstract
Exposure to extremely low frequency pulsed electromagnetic fields (ELF-PEMF) is supposed to simulate local EMF generated during mechanical stimulation of bone and may therefore be used to improve bone regeneration. This study aimed at optimizing the exposure strategy and investigating the underlying mechanisms of a 16 Hz ELF-PEMF, previously reported to boost osteoblast function. Comparing influences of daily continuous (30 min every 24 h) and intermittent (10 min every 8 h) exposure to the 16 Hz ELF-PEMF on osteoprogenitor cells revealed that the intermittent exposure strategy enhanced the 16 Hz ELF-PEMF effects regarding cell numbers and osteogenic function. Gene expression of piezo 1 and related Ca2+ influx were significantly increased in SCP-1 cells with the daily intermittent exposure. Pharmacological inhibition of piezo 1 with Dooku 1 largely abolished the positive effect of the 16 Hz ELF-PEMF exposure on osteogenic maturation of SCP-1 cells. In summary, the intermittent exposure strategy enhanced the positive effects of 16 Hz continuous ELF-PEMF exposure in terms of cell viability and osteogenesis. This effect was shown to be mediated by an increased expression of piezo 1 and related Ca2+ influx. Thus, the intermittent exposure strategy is a promising way to further optimize the therapeutic effects of the 16 Hz ELF-PEMF regarding fracture healing or osteoporosis.
Collapse
Affiliation(s)
- Yangmengfan Chen
- Siegfried Weller Institute at the BG Trauma Center Tübingen, Department of Trauma and Reconstructive Surgery, University of Tübingen, Schnarrenbergstr. 95, D-72076 Tübingen, Germany
| | - Benedikt J. Braun
- Siegfried Weller Institute at the BG Trauma Center Tübingen, Department of Trauma and Reconstructive Surgery, University of Tübingen, Schnarrenbergstr. 95, D-72076 Tübingen, Germany
| | - Maximilian M. Menger
- Siegfried Weller Institute at the BG Trauma Center Tübingen, Department of Trauma and Reconstructive Surgery, University of Tübingen, Schnarrenbergstr. 95, D-72076 Tübingen, Germany
| | - Michael Ronniger
- Sachtleben GmbH, Haus Spectrum am UKE, Martinistraße 64, D-20251 Hamburg, Germany
| | - Karsten Falldorf
- Sachtleben GmbH, Haus Spectrum am UKE, Martinistraße 64, D-20251 Hamburg, Germany
| | - Tina Histing
- Siegfried Weller Institute at the BG Trauma Center Tübingen, Department of Trauma and Reconstructive Surgery, University of Tübingen, Schnarrenbergstr. 95, D-72076 Tübingen, Germany
| | - Andreas K. Nussler
- Siegfried Weller Institute at the BG Trauma Center Tübingen, Department of Trauma and Reconstructive Surgery, University of Tübingen, Schnarrenbergstr. 95, D-72076 Tübingen, Germany
| | - Sabrina Ehnert
- Siegfried Weller Institute at the BG Trauma Center Tübingen, Department of Trauma and Reconstructive Surgery, University of Tübingen, Schnarrenbergstr. 95, D-72076 Tübingen, Germany
| |
Collapse
|
19
|
Abed T, Ganser K, Eckert F, Stransky N, Huber SM. Ion channels as molecular targets of glioblastoma electrotherapy. Front Cell Neurosci 2023; 17:1133984. [PMID: 37006466 PMCID: PMC10064067 DOI: 10.3389/fncel.2023.1133984] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Accepted: 02/10/2023] [Indexed: 03/19/2023] Open
Abstract
Therapies with weak, non-ionizing electromagnetic fields comprise FDA-approved treatments such as Tumor Treating Fields (TTFields) that are used for adjuvant therapy of glioblastoma. In vitro data and animal models suggest a variety of biological TTFields effects. In particular, effects ranging from direct tumoricidal, radio- or chemotherapy-sensitizing, metastatic spread-inhibiting, up to immunostimulation have been described. Diverse underlying molecular mechanisms, such as dielectrophoresis of cellular compounds during cytokinesis, disturbing the formation of the spindle apparatus during mitosis, and perforating the plasma membrane have been proposed. Little attention, however, has been paid to molecular structures that are predestinated to percept electromagnetic fields-the voltage sensors of voltage-gated ion channels. The present review article briefly summarizes the mode of action of voltage sensing by ion channels. Moreover, it introduces into the perception of ultra-weak electric fields by specific organs of fishes with voltage-gated ion channels as key functional units therein. Finally, this article provides an overview of the published data on modulation of ion channel function by diverse external electromagnetic field protocols. Combined, these data strongly point to a function of voltage-gated ion channels as transducers between electricity and biology and, hence, to voltage-gated ion channels as primary targets of electrotherapy.
Collapse
Affiliation(s)
- Tayeb Abed
- Department of Radiation Oncology, University of Tübingen, Tübingen, Germany
| | - Katrin Ganser
- Department of Radiation Oncology, University of Tübingen, Tübingen, Germany
| | - Franziska Eckert
- Department of Radiation Oncology, University of Tübingen, Tübingen, Germany
- Department of Radiation Oncology, Medical University Vienna, Vienna, Austria
| | - Nicolai Stransky
- Department of Radiation Oncology, University of Tübingen, Tübingen, Germany
- Department of Pharmacology, Toxicology and Clinical Pharmacy, Institute of Pharmacy, University of Tübingen, Tübingen, Germany
| | - Stephan M. Huber
- Department of Radiation Oncology, University of Tübingen, Tübingen, Germany
| |
Collapse
|
20
|
Chen Y, Lu C, Shang X, Wu K, Chen K. Primary cilia: The central role in the electromagnetic field induced bone healing. Front Pharmacol 2022; 13:1062119. [DOI: 10.3389/fphar.2022.1062119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Accepted: 11/07/2022] [Indexed: 12/03/2022] Open
Abstract
Primary cilia have emerged as the cellular “antenna” that can receive and transduce extracellular chemical/physical signals, thus playing an important role in regulating cellular activities. Although the electromagnetic field (EMF) is an effective treatment for bone fractures since 1978, however, the detailed mechanisms leading to such positive effects are still unclear. Primary cilia may play a central role in receiving EMF signals, translating physical signals into biochemical information, and initiating various signalingsignaling pathways to transduce signals into the nucleus. In this review, we elucidated the process of bone healing, the structure, and function of primary cilia, as well as the application and mechanism of EMF in treating fracture healing. To comprehensively understand the process of bone healing, we used bioinformatics to analyze the molecular change and associated the results with other studies. Moreover, this review summarizedsummarized some limitations in EMFs-related research and provides an outlook for ongoing studies. In conclusion, this review illustrated the primary cilia and related molecular mechanisms in the EMF-induced bone healing process, and it may shed light on future research.
Collapse
|
21
|
Sadeghian-Nodoushan F, Nikukar H, Soleimani M, Jalali- Jahromi A, Hosseinzadeh S, Khojasteh A. A smart magnetic hydrogel containing exosome promotes osteogenic commitment of human adipose-derived mesenchymal stem cells. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2022; 25:1123-1131. [PMID: 36246059 PMCID: PMC9526893 DOI: 10.22038/ijbms.2022.64682.14237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Accepted: 08/21/2022] [Indexed: 11/05/2022]
Abstract
Objectives Exosomes, as nano-sized extracellular vehicles acting as cell-to-cell communicators, are novel promising therapeutics in the area of bone tissue engineering. Moreover, magnetic nanoparticles, whose integration with other appropriate components is viewed as an intriguing approach to strengthen bone tissue engineering efficacy. We investigated the effect of magnetic enriched with exosomes on osteogenic differentiation. Materials and Methods Exosomes were isolated from human adipose-derived mesenchymal stem cells by Exo-spin™ kit (MSC-EX). Alginate (Alg) scaffold containing 1% (w/w) cobalt ferrite nanoparticles (CoFe2O4) was produced. MSC-EX were gently loaded onto Alg and Alg-cobalt ferrite (Alg-CF) scaffolds yielding Alg-EX and Alg-CF-EX scaffolds. The effects of MSC-Ex and magnetic hydrogel composite under an external static magnetic field (SMF) on proliferation and differentiation of MSCs were evaluated by alkaline phosphatase (ALP) activity measurement, alizarin red staining, and energy dispersive X-ray (EDX) analysis. Results Our results showed that Alg and Alg-CF scaffolds were not only cytotoxic but also supported AdMSCs proliferation. MSC-EX loading of the scaffolds enhanced AdMSCs proliferation significantly. According to the results, Alg-CF-EX scaffolds under magnetic stimulation exhibited the most potent effect on osteogenic differentiation of cultured AdMSCs as evidenced by higher ALP activity and mineralization. Conclusion We provided evidence that the combination of Alg hydrogel, CFNPs, and MSC-EX resulted in the construction of a bone tissue-engineering scaffold that highly supports the osteogenic commitment of MSCs.
Collapse
Affiliation(s)
- Fatemeh Sadeghian-Nodoushan
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Habib Nikukar
- Research and Clinical Center for Infertility, Yazd Reproductive Sciences Institute, Shahid Sadoughi University of Medical Sciences, Yazd, Iran,Medical Nanotechnology and Tissue Engineering Research Center, Yazd Reproductive Sciences Institute, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Masoud Soleimani
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Azarmidokht Jalali- Jahromi
- Medical Nanotechnology and Tissue Engineering Research Center, Yazd Reproductive Sciences Institute, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Simzar Hosseinzadeh
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran,Medical Nanotechnology and Tissue Engineering Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran,Corresponding authors: Simzar Hosseinzadeh. Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran. Tel: +98-21-88666136; ; Arash Khojasteh. Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran. Tel: +98-21-88666136;
| | - Arash Khojasteh
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran,Department of Health and Medical Sciences, University of Antwerp, Antwerp, Belgium ,Corresponding authors: Simzar Hosseinzadeh. Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran. Tel: +98-21-88666136; ; Arash Khojasteh. Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran. Tel: +98-21-88666136;
| |
Collapse
|
22
|
Osteogenic effect of electromagnetic fields on stem cells derived from rat bone marrow cultured in osteogenic medium versus conditioned medium in vitro. Cell Tissue Bank 2022; 24:317-328. [PMID: 36042070 DOI: 10.1007/s10561-022-10034-4] [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: 04/18/2022] [Accepted: 08/03/2022] [Indexed: 11/02/2022]
Abstract
OBJECTIVES This study assessed possible osteogenic differentiation caused by electromagnetic fields (EMF) on rat bone-marrow-derived stem cells (rBMSCs) cultured in osteogenic medium (OM) or in human adipose-stem cell-conditioned medium (hADSC-CM). MATERIALS AND METHODS The rBMSCs were divided into negative and positive control groups, cultured in α-MEM plus 10% FBS or OM respectively. CM and CM + EMF groups, cultured cells in hADSCs-CM or exposed to EMF (50 Hz, 1 mT) for 30 min/day plus hADSCs-CM, respectively. Cells from the OM + EMF were simultaneously cultured in OM and exposed to EMF. Osteogenesis was investigated through alkaline phosphatase activity, alizarin red staining and real-time PCR. RESULTS A meaningfully higher level of ALP activity was observed in the OM + EMF group compared to the other groups. There was a considerable increase in Runx2 expression in the CM + EMF group compared to the positive control and CM groups and a significant increase in Runx2 expression in the OM + EMF in comparison with all other groups after 21 days. Runx2 expression increased significantly in the CM, CM + EMF and positive control groups on day 21 compared to the same groups on day 14. From days 14-21, Ocn expression increased in the CM and CM + EMF groups, but both groups showed a significant decrease compared to the positive controls. CM and EMF had no effect on Ocn expression. On day 21, Ocn expression was significantly higher in the OM + EMF group than in the positive control group. CONCLUSION The synergistic effect of EMF and OM increased the expression of Runx2 and Ocn in rBMSCs.
Collapse
|
23
|
Lang S, Ma J, Gong S, Wang Y, Dong B, Ma X. Pulse Electromagnetic Field for Treating Postmenopausal Osteoporosis: A Systematic Review and Meta-Analysis of Randomized Controlled Trials. Bioelectromagnetics 2022; 43:381-393. [PMID: 35864717 DOI: 10.1002/bem.22419] [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: 03/30/2022] [Revised: 06/25/2022] [Accepted: 06/30/2022] [Indexed: 11/09/2022]
Abstract
Postmenopausal osteoporosis is a type of chronic disease with high morbidity and high economic burden. Due to the adverse effects of long-term drug therapy, physical therapy, such as pulsed electromagnetic fields (PEMF), is widely implemented in clinical practice. Therefore, we first conducted the meta-analysis on the efficacy and safety of PEMF in the treatment of postmenopausal osteoporosis. We searched eight databases to acquire potentially eligible studies. Outcome indicators include bone mineral density (BMD), visual analogue scale (VAS), biochemical markers of alkaline phosphatase (ALP), osteocalcin, bone-specific alkaline phosphatase (BSAP), type I collagen carboxy-terminal peptide (CTX), and adverse events. The results showed that a total of 19 studies (1303 patients) were retrieved from eight databases. Compared with conventional medications, PEMF combined with conventional medications significantly increased BMD of lumbar vertebra, femoral, Ward's triangle, bone-specific biochemical indicators of ALP, BSAP, and osteocalcin, and relieved pain. However, The incidence of adverse events was not statistically significant between PEMF combined with conventional medications and conventional medications alone. Compared with conventional medications, PEMF significantly increased the BMD of the femur and reduced the degree of pain, but there was no statistical difference in the BMD of the lumbar spine between PEMF and placebo. Except osteocalcin, BSAP, CTX, and ALP showed no significant difference. In view of its efficacy and safety, PEMF intervention can be considered as a potentially effective complementary therapy for postmenopausal women with osteoporosis. © 2022 Bioelectromagnetics Society.
Collapse
Affiliation(s)
- Shuang Lang
- Tianjin Key Laboratory of Orthopedic Biomechanics and Medical Engineering, Orthopedic Research Institute, Tianjin Hospital, Tianjin, China.,Orthopaedics Institute, Tianjin Hospital, Tianjin, China.,Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Jianxiong Ma
- Tianjin Key Laboratory of Orthopedic Biomechanics and Medical Engineering, Orthopedic Research Institute, Tianjin Hospital, Tianjin, China.,Orthopaedics Institute, Tianjin Hospital, Tianjin, China
| | - Shuwei Gong
- Tianjin Key Laboratory of Orthopedic Biomechanics and Medical Engineering, Orthopedic Research Institute, Tianjin Hospital, Tianjin, China.,Orthopaedics Institute, Tianjin Hospital, Tianjin, China.,Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yan Wang
- Tianjin Key Laboratory of Orthopedic Biomechanics and Medical Engineering, Orthopedic Research Institute, Tianjin Hospital, Tianjin, China
| | - Benchao Dong
- Tianjin Key Laboratory of Orthopedic Biomechanics and Medical Engineering, Orthopedic Research Institute, Tianjin Hospital, Tianjin, China
| | - Xinlong Ma
- Tianjin Key Laboratory of Orthopedic Biomechanics and Medical Engineering, Orthopedic Research Institute, Tianjin Hospital, Tianjin, China.,Orthopaedics Institute, Tianjin Hospital, Tianjin, China
| |
Collapse
|
24
|
Guillot-Ferriols M, Lanceros-Méndez S, Gómez Ribelles JL, Gallego Ferrer G. Electrical stimulation: Effective cue to direct osteogenic differentiation of mesenchymal stem cells? BIOMATERIALS ADVANCES 2022; 138:212918. [PMID: 35913228 DOI: 10.1016/j.bioadv.2022.212918] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 05/02/2022] [Accepted: 05/20/2022] [Indexed: 06/15/2023]
Abstract
Mesenchymal stem cells (MSCs) play a major role in bone tissue engineering (BTE) thanks to their capacity for osteogenic differentiation and being easily available. In vivo, MSCs are exposed to an electroactive microenvironment in the bone niche, which has piezoelectric properties. The correlation between the electrically active milieu and bone's ability to adapt to mechanical stress and self-regenerate has led to using electrical stimulation (ES) as physical cue to direct MSCs differentiation towards the osteogenic lineage in BTE. This review summarizes the different techniques to electrically stimulate MSCs to induce their osteoblastogenesis in vitro, including general electrical stimulation and substrate mediated stimulation by means of conductive or piezoelectric cell culture supports. Several aspects are covered, including stimulation parameters, treatment times and cell culture media to summarize the best conditions for inducing MSCs osteogenic commitment by electrical stimulation, from a critical point of view. Electrical stimulation activates different signaling pathways, including bone morphogenetic protein (BMP) Smad-dependent or independent, regulated by mitogen activated protein kinases (MAPK), extracellular signal-regulated kinases (ERK) and p38. The roles of voltage gate calcium channels (VGCC) and integrins are also highlighted according to their application technique and parameters, mainly converging in the expression of RUNX2, the master regulator of the osteogenic differentiation pathway. Despite the evident lack of homogeneity in the approaches used, the ever-increasing scientific evidence confirms ES potential as an osteoinductive cue, mimicking aspects of the in vivo microenvironment and moving one step forward to the translation of this approach into clinic.
Collapse
Affiliation(s)
- M Guillot-Ferriols
- Centre for Biomaterials and Tissue Engineering (CBIT), Universitat Politècnica de València, 46022 Valencia, Spain; Biomedical Research Networking Centre on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Valencia, Spain.
| | - S Lanceros-Méndez
- Centre of Physics of Minho and Porto Universities, Universidade do Minho, 4710-058 Braga, Portugal; BCMaterials, Basque Centre for Materials, Applications and Nanostructures, UPV/EHU Science Park, 48940 Leioa, Spain; IKERBASQUE, Basque Foundation for Science, 48009 Bilbao, Spain
| | - J L Gómez Ribelles
- Centre for Biomaterials and Tissue Engineering (CBIT), Universitat Politècnica de València, 46022 Valencia, Spain; Biomedical Research Networking Centre on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Valencia, Spain
| | - G Gallego Ferrer
- Centre for Biomaterials and Tissue Engineering (CBIT), Universitat Politècnica de València, 46022 Valencia, Spain; Biomedical Research Networking Centre on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Valencia, Spain
| |
Collapse
|
25
|
Chen L, Cheng G, Meng S, Ding Y. Collagen Membrane Derived from Fish Scales for Application in Bone Tissue Engineering. Polymers (Basel) 2022; 14:polym14132532. [PMID: 35808577 PMCID: PMC9269230 DOI: 10.3390/polym14132532] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2022] [Revised: 06/03/2022] [Accepted: 06/10/2022] [Indexed: 02/06/2023] Open
Abstract
Guided tissue/bone regeneration (GTR/GBR) is currently the main treatment for alveolar bone regeneration. The commonly used barrier membranes in GTR/GBR are collagen membranes from mammals such as porcine or cattle. Fish collagen is being explored as a potential substitute for mammalian collagen due to its low cost, no zoonotic risk, and lack of religious constraints. Fish scale is a multi-layer natural collagen composite with high mechanical strength, but its biomedical application is limited due to the low denaturation temperature of fish collagen. In this study, a fish scale collagen membrane with a high denaturation temperature of 79.5 °C was prepared using an improved method based on preserving the basic shape of fish scales. The fish scale collagen membrane was mainly composed of type I collagen and hydroxyapatite, in which the weight ratios of water, organic matter, and inorganic matter were 20.7%, 56.9%, and 22.4%, respectively. Compared to the Bio-Gide® membrane (BG) commonly used in the GTR/GBR, fish scale collagen membrane showed good cytocompatibility and could promote late osteogenic differentiation of cells. In conclusion, the collagen membrane prepared from fish scales had good thermal stability, cytocompatibility, and osteogenic activity, which showed potential for bone tissue engineering applications.
Collapse
Affiliation(s)
- Liang Chen
- National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China; (L.C.); (G.C.); (S.M.)
- State Key Laboratory of Oral Diseases, Sichuan University, Chengdu 610041, China
- Department of Periodontology, West China College of Stomatology, Sichuan University, Chengdu 610041, China
- Department of Periodontology, Peking University School and Hospital of Stomatology, Beijing 100081, China
| | - Guoping Cheng
- National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China; (L.C.); (G.C.); (S.M.)
- State Key Laboratory of Oral Diseases, Sichuan University, Chengdu 610041, China
- Department of Periodontology, West China College of Stomatology, Sichuan University, Chengdu 610041, China
| | - Shu Meng
- National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China; (L.C.); (G.C.); (S.M.)
- State Key Laboratory of Oral Diseases, Sichuan University, Chengdu 610041, China
- Department of Periodontology, West China College of Stomatology, Sichuan University, Chengdu 610041, China
| | - Yi Ding
- National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China; (L.C.); (G.C.); (S.M.)
- State Key Laboratory of Oral Diseases, Sichuan University, Chengdu 610041, China
- Department of Periodontology, West China College of Stomatology, Sichuan University, Chengdu 610041, China
- Correspondence:
| |
Collapse
|
26
|
He WF, Qin R, Gao YH, Zhou J, Wei JJ, Liu J, Hou XF, Ma HP, Xian CJ, Li XY, Chen KM. The interdependent relationship between the nitric oxide signaling pathway and primary cilia in pulse electromagnetic field-stimulated osteoblastic differentiation. FASEB J 2022; 36:e22376. [PMID: 35616355 DOI: 10.1096/fj.202101577rr] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 05/06/2022] [Accepted: 05/13/2022] [Indexed: 11/11/2022]
Abstract
Pulsed electromagnetic fields (PEMFs) have long been recognized being safe and effective in treating bone fracture nonunion and osteoporosis. However, the mechanism of osteogenic action of PEMFs is still unclear. While primary cilia are reported to be a sensory organelle for PEMFs, and nitric oxide (NO) plays an indispensable role in osteogenic effect of PEMFs, the relationship between NO and primary cilia is unknown. In this study, effects of treatment with 50 Hz 0.6 mT PEMFs on osteogenic differentiation and mineralization, NO secretion, and ciliary location of specific proteins were examined in rat calvarial osteoblasts (ROBs) with normal or abrogated primary cilia. It was found that PEMFs stimulated the osteogenic differentiation by activating the NOS/NO/sGC/cGMP/PKG signaling pathway, which need the existence of primary cilia. All components of the signaling pathway including iNOS, eNOS, sGC, PKG-1, and PKG-2 were localized to primary cilia, and eNOS was phosphorylated inside the primary cilia. Besides, primary cilia were elongated significantly by PEMF treatment and changed dynamically with the activation NO/cGMP pathway. When the pathway was blocked by L-NAME, PEMFs could no longer elongate the primary cilia and stimulate the osteoblastic differentiation. Thus, this study for the first time observed activation of the NO/cGMP signaling pathway in ciliary compartment of osteoblasts, and PEMFs could not stimulate the osteoblastic differentiation if the NO signaling pathway was blocked or the ciliogenesis was inhibited. Our findings indicate the interdependent relationship between NO and primary cilia in the PEMF-promoted osteogenesis.
Collapse
Affiliation(s)
- Wen-Fang He
- Fundamental Medical Science Research Laboratories, The 940th Hospital of Joint Logistic Support Force, People's Liberation Army of China, Lanzhou, P. R. China.,Department of Bioengineering, School of Life Science and Engineering, Lanzhou University of Technology, Lanzhou, P. R. China.,Key Laboratory of Stem Cells and Gene Drugs of Gansu Province, Lanzhou, P. R. China
| | - Rong Qin
- Fundamental Medical Science Research Laboratories, The 940th Hospital of Joint Logistic Support Force, People's Liberation Army of China, Lanzhou, P. R. China
| | - Yu-Hai Gao
- Fundamental Medical Science Research Laboratories, The 940th Hospital of Joint Logistic Support Force, People's Liberation Army of China, Lanzhou, P. R. China
| | - Jian Zhou
- Fundamental Medical Science Research Laboratories, The 940th Hospital of Joint Logistic Support Force, People's Liberation Army of China, Lanzhou, P. R. China
| | - Juan-Juan Wei
- Fundamental Medical Science Research Laboratories, The 940th Hospital of Joint Logistic Support Force, People's Liberation Army of China, Lanzhou, P. R. China
| | - Jing Liu
- Fundamental Medical Science Research Laboratories, The 940th Hospital of Joint Logistic Support Force, People's Liberation Army of China, Lanzhou, P. R. China
| | - Xue-Feng Hou
- Fundamental Medical Science Research Laboratories, The 940th Hospital of Joint Logistic Support Force, People's Liberation Army of China, Lanzhou, P. R. China
| | - Hui-Ping Ma
- Fundamental Medical Science Research Laboratories, The 940th Hospital of Joint Logistic Support Force, People's Liberation Army of China, Lanzhou, P. R. China
| | - Cory J Xian
- UniSA Clinical and Health Sciences, University of South Australia, Adelaide, South Australia, Australia
| | - Xue-Yan Li
- Department of Bioengineering, School of Life Science and Engineering, Lanzhou University of Technology, Lanzhou, P. R. China
| | - Ke-Ming Chen
- Fundamental Medical Science Research Laboratories, The 940th Hospital of Joint Logistic Support Force, People's Liberation Army of China, Lanzhou, P. R. China.,Key Laboratory of Stem Cells and Gene Drugs of Gansu Province, Lanzhou, P. R. China
| |
Collapse
|
27
|
Volz M, Wyse-Sookoo KR, Travascio F, Huang CY, Best TM. MECHANOBIOLOGICAL APPROACHES FOR STIMULATING CHONDROGENESIS OF STEM CELLS. Stem Cells Dev 2022; 31:460-487. [PMID: 35615879 DOI: 10.1089/scd.2022.0049] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Chondrogenesis is the process of differentiation of stem cells into mature chondrocytes. Such a process consists of chemical, functional, and structural changes which are initiated and mediated by the host environment of the cells. To date, the mechanobiology of chondrogenesis has not been fully elucidated. Hence, experimental activity is focused on recreating specific environmental conditions for stimulating chondrogenesis, and to look for a mechanistic interpretation of the mechanobiological response of cells in the cartilaginous tissues. There are a large number of studies on the topic that vary considerably in their experimental protocols used for providing environmental cues to cells for differentiation, making generalizable conclusions difficult to ascertain. The main objective of this contribution is to review the mechanobiological stimulation of stem cell chondrogenesis and methodological approaches utilized to date to promote chondrogenesis of stem cells in-vitro. In-vivo models will also be explored, but this area is currently limited. An overview of the experimental approaches used by different research groups may help the development of unified testing methods that could be used to overcome existing knowledge gaps, leading to an accelerated translation of experimental findings to clinical practice.
Collapse
Affiliation(s)
- Mallory Volz
- University of Miami, 5452, Biomedical Engineering, Coral Gables, Florida, United States;
| | | | - Francesco Travascio
- University of Miami, 5452, Mechanical and Aerospace Engineering, 1251 Memorial Drive, MEB 217B, Coral Gables, Florida, United States, 33146;
| | - Chun-Yuh Huang
- University of Miami, 5452, Biomedical Engineering, Coral Gables, Florida, United States;
| | - Thomas M Best
- University of Miami Miller School of Medicine, 12235, School of Medicine, Miami, Florida, United States;
| |
Collapse
|
28
|
Ye AF, Liu XC, Chen LJ, Xia YP, Yang XB, Sun WJ. Endogenous Ca 2+ release was involved in 50-Hz MF-induced proliferation via Akt-SK1 signal cascade in human amniotic epithelial cells. Electromagn Biol Med 2022; 41:142-151. [PMID: 35129008 DOI: 10.1080/15368378.2022.2031211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
The mechanism underlying the biological effects caused by an extremely low-frequency electromagnetic field (ELF-EMF) is still unclear. Previously, we found that L-type calcium channel and sphingosine kinase 1 (SK1) were involved in 50-Hz MF exposure-induced cell proliferation. In the present study, the role of intracellular Ca2+ and signal molecules related to SK1 in cell proliferation induced by 50-Hz MF was investigated in human amniotic epithelial (FL) cells. Results showed that the intracellular Ca2+ chelator, BAPTA, could completely inhibit 50-Hz MF-induced cell proliferation, whereas NIF, the inhibitor of L-type calcium channel, only partly blocked it. When cells were cultured in calcium-free medium, MF exposure also increased intracellular Ca2+, activated SK1 and promoted cell proliferation although all of those increasing levels were lower than those in complete medium. Moreover, MF-activated SK1 could be completely inhibited by BAPTA, and MF-induced cell proliferation was abolished by SKI II, the specific inhibitor of SK1. Additionally, a 50-Hz MF exposure did not affect the activation of ERK and PKCα under the condition of calcium-free medium, but activated the Akt, which could be precluded entirely by BAPTA, but not be inhibited by NIF. Treatment of FL cells with LY294002, the inhibitor of Akt, could delete the MF-induced SK1 activation under the condition of calcium-free medium. Based on the data from the present experiment, it is concluded that endogenous Ca2+ release was involved in 50-Hz MF-induced cell proliferation via Akt-SK1 signal cascade.
Collapse
Affiliation(s)
- An-Fang Ye
- The First Affiliated Hospital, School of Public Health, Zhejiang University School of Medicine, Hangzhou, ZJ, China
| | - Xiao-Chen Liu
- The First Affiliated Hospital, School of Public Health, Zhejiang University School of Medicine, Hangzhou, ZJ, China
| | - Liang-Jing Chen
- Institute of Environmental Medicine, Zhejiang University School of Medicine, Hangzhou, ZJ, China.,Bioelectromagnetics Key Laboratory, Zhejiang University School of Medicine, Hangzhou, ZJ, China
| | - Yong-Peng Xia
- Institute of Environmental Medicine, Zhejiang University School of Medicine, Hangzhou, ZJ, China.,Shaoxing Shangyu Area Center for Disease Control and Prevention, Shaoxing, ZJ, China
| | - Xiao-Bo Yang
- Institute of Environmental Medicine, Zhejiang University School of Medicine, Hangzhou, ZJ, China.,Tianjin Institute of Environmental and Operational Medicine, Tianjin, China
| | - Wen-Jun Sun
- The First Affiliated Hospital, School of Public Health, Zhejiang University School of Medicine, Hangzhou, ZJ, China.,Institute of Environmental Medicine, Zhejiang University School of Medicine, Hangzhou, ZJ, China.,Bioelectromagnetics Key Laboratory, Zhejiang University School of Medicine, Hangzhou, ZJ, China
| |
Collapse
|
29
|
Zhu S, Li Y, Wang L, Huang J, Song K, Gan X, Xiang X, He C, Yang L. Pulsed electromagnetic fields may be effective for the management of primary osteoporosis: a systematic review and meta-analysis. IEEE Trans Neural Syst Rehabil Eng 2022; 30:321-328. [PMID: 35130160 DOI: 10.1109/tnsre.2022.3149483] [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: 11/06/2022]
Abstract
Little is known about the effect of pulsed electromagnetic fields (PEMFs) as an option for preventing osteoporosis. This study sought to investigate the effectiveness of PEMFs for the management of primary osteoporosis in older adults. We searched databases from the inception to date to target trials examining the effects of PEMFs compared to placebo or sham or other agents for the management of primary osteoporosis for a meta-analysis using random effects model. Eight trials including 411 participants were included. PEMFs was non-inferior to conventional pharmacological agents and exercise respectively in preventing the decline of Bone Mineral Density (BMD) at the lumbar (MD 8.76; CI -9.64 to 27.16 and MD 1.33; CI -2.73 to 5.39) and femur neck (MD 0.04; CI -1.09 to 1.16 and MD 1.50; CI -0.26 to 3.26), and significantly improving balance function measured by Berg Balance Scale (BBS) (MD 0.91; CI 0.32 to 1.49) and Timed Up and Go test (MD -3.61; CI -6.37 to -0.85), directly after intervention. The similar trends were observed in BMD and BBS at 12- and 24-weeks follow-up from baseline. PEMFs had positive effects non-inferior to first-line treatment on BMD and better over placebo on balance function in older adults with primary osteoporosis, but with moderate to very low certainty evidence and short-term follow-ups. There is a need for high-quality randomised controlled trials evaluating PEMFs for the management of primary osteoporosis.
Collapse
|
30
|
Wang X, Zhao R, Wang J, Li X, Jin L, Liu W, Yang L, Zhu Y, Tan Z. 3D-printed tissue repair patch combining mechanical support and magnetism for controlled skeletal muscle regeneration. Biodes Manuf 2022. [DOI: 10.1007/s42242-021-00180-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
31
|
Li G, Chang B, He Y, Li Y, Liu J, Zhang Y, Hou Y, Xu B, Li X, Xu M, Ding X, Song W, Zhang Y. Orai1 mediated store-operated calcium entry contributing to MC3T3-E1 differentiation on titanium implant with micro/nano-textured topography. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2022; 133:112644. [DOI: 10.1016/j.msec.2022.112644] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 12/13/2021] [Accepted: 01/02/2022] [Indexed: 10/19/2022]
|
32
|
Kong Y, Duan J, Liu F, Han L, Li G, Sun C, Sang Y, Wang S, Yi F, Liu H. Regulation of stem cell fate using nanostructure-mediated physical signals. Chem Soc Rev 2021; 50:12828-12872. [PMID: 34661592 DOI: 10.1039/d1cs00572c] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
One of the major issues in tissue engineering is regulation of stem cell differentiation toward specific lineages. Unlike biological and chemical signals, physical signals with adjustable properties can be applied to stem cells in a timely and localized manner, thus making them a hot topic for research in the fields of biomaterials, tissue engineering, and cell biology. According to the signals sensed by cells, physical signals used for regulating stem cell fate can be classified into six categories: mechanical, light, thermal, electrical, acoustic, and magnetic. In most cases, external macroscopic physical fields cannot be used to modulate stem cell fate, as only the localized physical signals accepted by the surface receptors can regulate stem cell differentiation via nanoscale fibrin polysaccharide fibers. However, surface receptors related to certain kinds of physical signals are still unknown. Recently, significant progress has been made in the development of functional materials for energy conversion. Consequently, localized physical fields can be produced by absorbing energy from an external physical field and subsequently releasing another type of localized energy through functional nanostructures. Based on the above concepts, we propose a methodology that can be utilized for stem cell engineering and for the regulation of stem cell fate via nanostructure-mediated physical signals. In this review, the combined effect of various approaches and mechanisms of physical signals provides a perspective on stem cell fate promotion by nanostructure-mediated physical signals. We expect that this review will aid the development of remote-controlled and wireless platforms to physically guide stem cell differentiation both in vitro and in vivo, using optimized stimulation parameters and mechanistic investigations while driving the progress of research in the fields of materials science, cell biology, and clinical research.
Collapse
Affiliation(s)
- Ying Kong
- State Key Laboratory of Crystal Materials, Shandong University, Jinan, 250100, China.
| | - Jiazhi Duan
- State Key Laboratory of Crystal Materials, Shandong University, Jinan, 250100, China.
| | - Feng Liu
- State Key Laboratory of Crystal Materials, Shandong University, Jinan, 250100, China.
| | - Lin Han
- Institute of Marine Science and Technology, Shandong University, Qingdao, 266200, China.
| | - Gang Li
- Neurological Surgery, Qilu Hospital of Shandong University, Jinan, 250012, China
| | - Chunhui Sun
- Collaborative Innovation Center of Technology and Equipment for Biological Diagnosis and Therapy in Universities of Shandong, Institute for Advanced Interdisciplinary Research (iAIR), University of Jinan, Jinan, 250022, China
| | - Yuanhua Sang
- State Key Laboratory of Crystal Materials, Shandong University, Jinan, 250100, China.
| | - Shuhua Wang
- State Key Laboratory of Crystal Materials, Shandong University, Jinan, 250100, China.
| | - Fan Yi
- The Key Laboratory of Infection and Immunity of Shandong Province, Department of Pharmacology, School of Basic Medical Science, Shandong University, Jinan, 250012, China.
| | - Hong Liu
- State Key Laboratory of Crystal Materials, Shandong University, Jinan, 250100, China. .,Collaborative Innovation Center of Technology and Equipment for Biological Diagnosis and Therapy in Universities of Shandong, Institute for Advanced Interdisciplinary Research (iAIR), University of Jinan, Jinan, 250022, China
| |
Collapse
|
33
|
Dhawan U, Jaffery H, Salmeron-Sanchez M, Dalby MJ. An ossifying landscape: materials and growth factor strategies for osteogenic signalling and bone regeneration. Curr Opin Biotechnol 2021; 73:355-363. [PMID: 34735985 DOI: 10.1016/j.copbio.2021.10.010] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 10/06/2021] [Accepted: 10/07/2021] [Indexed: 12/25/2022]
Abstract
Breakthroughs in our understanding of the complex interplay between cellular nanoenvironment and biomolecular signalling pathways are facilitating development of targeted osteogenic platforms. As critical biomolecules for osteogenesis, growth factors stimulate osteogenesis by activating key genes and transcription factors. The first half of this review presents emerging interconnectedness and recent discoveries of osteogenic signalling pathways initiating from growth factors for example, bone morphogenetic protein 2 (BMP-2). To complement this, the second half of review proposes a number of strategies to induce osteogenesis which include metallic, organic implants, nanotopological environments as well as growth factor immobilization techniques. The drawbacks of traditional osteogenic implants and how these have been overcome by biomedical engineers in the recent years without producing side-effects have also been summarized.
Collapse
Affiliation(s)
- Udesh Dhawan
- Centre for the Cellular Microenvironment, Division of Biomedical Engineering, School of Engineering, University of Glasgow, Glasgow, G12 8LT, UK
| | - Hussain Jaffery
- Centre for the Cellular Microenvironment, Institute of Molecular, Cell and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, G12 8QQ, UK
| | - Manuel Salmeron-Sanchez
- Centre for the Cellular Microenvironment, Division of Biomedical Engineering, School of Engineering, University of Glasgow, Glasgow, G12 8LT, UK
| | - Matthew J Dalby
- Centre for the Cellular Microenvironment, Institute of Molecular, Cell and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, G12 8QQ, UK.
| |
Collapse
|
34
|
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: 1.0] [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
|
35
|
Suryani L, Foo JKR, Cardilla A, Dong Y, Muthukumaran P, Hassanbhai A, Wen F, Simon DT, Iandolo D, Yu N, Ng KW, Teoh SH. Effects of Pulsed Electromagnetic Field Intensity on Mesenchymal Stem Cells. Bioelectricity 2021. [DOI: 10.1089/bioe.2021.0002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Affiliation(s)
- Luvita Suryani
- School of Chemical and Biomedical Engineering, College of Engineering, Nanyang Technological University, Singapore, Singapore
| | - Jyong Kiat Reuben Foo
- School of Chemical and Biomedical Engineering, College of Engineering, Nanyang Technological University, Singapore, Singapore
| | - Angelysia Cardilla
- School of Chemical and Biomedical Engineering, College of Engineering, Nanyang Technological University, Singapore, Singapore
| | - Yibing Dong
- School of Materials Science and Engineering, Nanyang Technological University, Singapore, Singapore
| | - Padmalosini Muthukumaran
- School of Chemical and Biomedical Engineering, College of Engineering, Nanyang Technological University, Singapore, Singapore
| | - Ammar Hassanbhai
- School of Chemical and Biomedical Engineering, College of Engineering, Nanyang Technological University, Singapore, Singapore
| | - Feng Wen
- School of Chemical and Biomedical Engineering, College of Engineering, Nanyang Technological University, Singapore, Singapore
| | - Daniel T. Simon
- Laboratory of Organic Electronics, Department of Science and Technology, Linköping University, Norrköping, Sweden
| | - Donata Iandolo
- Laboratory of Organic Electronics, Department of Science and Technology, Linköping University, Norrköping, Sweden
- UMR5510 MATEIS, CNRS, INSA-Lyon, University of Lyon, Lyon, France
- Mines Saint-Etienne, INSERM, U1059 SAINBIOSE, Saint-Étienne, France
| | - Na Yu
- National Dental Centre Singapore, Singapore, Singapore
- Duke-NUS Medical School, Singapore, Singapore
| | - Kee Woei Ng
- School of Materials Science and Engineering, Nanyang Technological University, Singapore, Singapore
- Environmental Chemistry & Materials Centre, Nanyang Environment and Water Research Institute (NEWRI), Nanyang Technological University, Singapore, Singapore
- Center for Nanotechnology and Nanotoxicology, Harvard T.H. Chan School of Public Health, Harvard University, Boston, Massachusetts, USA
| | - Swee-Hin Teoh
- School of Chemical and Biomedical Engineering, College of Engineering, Nanyang Technological University, Singapore, Singapore
- Lee Kong Chian School of Medicine, Singapore, Singapore
| |
Collapse
|
36
|
Adler D, Shapira Z, Weiss S, Shainberg A, Katz A. Weak Electromagnetic Fields Accelerate Fusion of Myoblasts. Int J Mol Sci 2021; 22:ijms22094407. [PMID: 33922487 PMCID: PMC8122904 DOI: 10.3390/ijms22094407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 04/21/2021] [Accepted: 04/21/2021] [Indexed: 11/28/2022] Open
Abstract
Weak electromagnetic fields (WEF) alter Ca2+ handling in skeletal muscle myotubes. Owing to the involvement of Ca2+ in muscle development, we investigated whether WEF affects fusion of myoblasts in culture. Rat primary myoblast cultures were exposed to WEF (1.75 µT, 16 Hz) for up to six days. Under control conditions, cell fusion and creatine kinase (CK) activity increased in parallel and peaked at 4–6 days. WEF enhanced the extent of fusion after one and two days (by ~40%) vs. control, but not thereafter. Exposure to WEF also enhanced CK activity after two days (almost four-fold), but not afterwards. Incorporation of 3H-thymidine into DNA was enhanced by one-day exposure to WEF (~40%), indicating increased cell replication. Using the potentiometric fluorescent dye di-8-ANEPPS, we found that exposure of cells to 150 mM KCl resulted in depolarization of the cell membrane. However, prior exposure of cells to WEF for one day followed by addition of KCl resulted in hyperpolarization of the cell membrane. Acute exposure of cells to WEF also resulted in hyperpolarization of the cell membrane. Twenty-four hour incubation of myoblasts with gambogic acid, an inhibitor of the inward rectifying K+ channel 2.1 (Kir2.1), did not affect cell fusion, WEF-mediated acceleration of fusion or hyperpolarization. These data demonstrate that WEF accelerates fusion of myoblasts, resulting in myotube formation. The WEF effect is associated with hyperpolarization but WEF does not appear to mediate its effects on fusion by activating Kir2.1 channels.
Collapse
Affiliation(s)
- Dana Adler
- Faculty of Life Sciences, Bar Ilan University, Ramat Gan 52900, Israel; (D.A.); (A.S.)
| | - Zehavit Shapira
- Department of Physics, Bar Ilan University, Ramat Gan 52900, Israel; (Z.S.); (S.W.)
| | - Shimon Weiss
- Department of Physics, Bar Ilan University, Ramat Gan 52900, Israel; (Z.S.); (S.W.)
- Department of Chemistry and Biochemistry, University of California, Los Angeles, CA 90095, USA
| | - Asher Shainberg
- Faculty of Life Sciences, Bar Ilan University, Ramat Gan 52900, Israel; (D.A.); (A.S.)
| | - Abram Katz
- Åstrand Laboratory of Work Physiology, The Swedish School of Sport and Health Sciences, GIH, Box 5626, SE-114 86 Stockholm, Sweden
- Correspondence:
| |
Collapse
|
37
|
Tabatabai TS, Haji Ghasem Kashani M, Maskani R, Nasiri M, Nabavi Amri SA, Atashi A, Bitaraf FS. Synergic effects of extremely low-frequency electromagnetic field and betaine on in vitro osteogenic differentiation of human adipose tissue-derived mesenchymal stem cells. In Vitro Cell Dev Biol Anim 2021; 57:468-476. [PMID: 33770338 DOI: 10.1007/s11626-021-00558-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Accepted: 02/25/2021] [Indexed: 11/30/2022]
Abstract
Human adipose tissue-derived mesenchymal stem cells (hADSCs) due to easy extraction, relative abundance, in vitro expansion and differentiation potential, frozen storage capability, and ability to secrete cytokines, compared to other stem cells, are appropriate candidate in regenerative medicine. Extremely low-frequency electromagnetic fields (ELF-EMF) and betaine are two safe factors in bone lesions repair. This study was designed to assess the osteogenic differentiation potential of these factors on hADSCs. The samples were collected from women undergoing liposuction after obtaining written consent. The hADSCs were extracted and treated with osteogenesis differentiation medium (OD) as the positive control, with OD and betaine (BET group), with OD and EMF (EMF group), and with OD and betaine and EMF (BET+EMF group) for 21 d; the negative control consisted of cells without treatment. Betaine 10 mM and EMF with 50-Hz frequency, 1-mT intensity (8 h daily), and in the form of sinus wave were used. Osteogenic differentiation was evaluated by Alizarin Red staining, alkaline phosphatase activity, calcium deposition, and real-time PCR. A significant increase in calcium deposition in the BET+EMF group was observed compared to the other groups. The activity of alkaline phosphatase in the positive control and BET groups was increased significantly compared to EMF and BET + EMF groups and a significant increase of this enzyme activity in the BET + EMF compared to EMF group was observed. The expression of RUNX2 and OCN genes in the EMF-treated groups were significantly reduced compared to the non-EMF-treated groups, and BET+EMF showed a significant increase of RUNX2 gene expression as compared the EMF group. The ELF-EMF leads to a decrease in the osteogenic differentiation and the expression RUNX2 and OCN genes in hADSCs. But osteogenic differentiation and RUNX2 gene expression were increased post-induction by betaine. The synergic effect of betaine and EMF on the osteogenic differentiation and related genes expression of hADSCs was higher than EMF.
Collapse
Affiliation(s)
- Tayebeh Sadat Tabatabai
- Department of Cellular and Molecular Biology, School of Biology and Institute of Biological Sciences, Damghan University, Damghan, Iran
| | - Maryam Haji Ghasem Kashani
- Department of Cellular and Molecular Biology, School of Biology and Institute of Biological Sciences, Damghan University, Damghan, Iran.
| | - Reza Maskani
- Department of Medical Sciences, School of Paramedical, Shahroud University of Medical Sciences, Shahroud, Iran
| | - Meysam Nasiri
- Department of Cellular and Molecular Biology, School of Biology and Institute of Biological Sciences, Damghan University, Damghan, Iran
| | | | - Amir Atashi
- Department of Medical Sciences, School of Paramedical, Shahroud University of Medical Sciences, Shahroud, Iran
| | - Fateme Sadat Bitaraf
- Department of Medical Biotechnology, School of Medicine, Shahroud University of Medical Sciences, Shahroud, Iran
| |
Collapse
|
38
|
Affiliation(s)
- Massimo Vassalli
- James Watt School of Engineering, University of Glasgow, Center for the Cellular Microenvironment, 70 University Avenue, School of Engineering, G12 8LT, Glasgow, UK
| |
Collapse
|
39
|
Janus 3D printed dynamic scaffolds for nanovibration-driven bone regeneration. Nat Commun 2021; 12:1031. [PMID: 33589620 PMCID: PMC7884435 DOI: 10.1038/s41467-021-21325-x] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Accepted: 01/13/2021] [Indexed: 12/12/2022] Open
Abstract
The application of physical stimuli to cell cultures has shown potential to modulate multiple cellular functions including migration, differentiation and survival. However, the relevance of these in vitro models to future potential extrapolation in vivo depends on whether stimuli can be applied “externally”, without invasive procedures. Here, we report on the fabrication and exploitation of dynamic additive-manufactured Janus scaffolds that are activated on-command via external application of ultrasounds, resulting in a mechanical nanovibration that is transmitted to the surrounding cells. Janus scaffolds were spontaneously formed via phase-segregation of biodegradable polycaprolactone (PCL) and polylactide (PLA) blends during the manufacturing process and behave as ultrasound transducers (acoustic to mechanical) where the PLA and PCL phases represent the active and backing materials, respectively. Remote stimulation of Janus scaffolds led to enhanced cell proliferation, matrix deposition and osteogenic differentiation of seeded human bone marrow derived stromal cells (hBMSCs) via formation and activation of voltage-gated calcium ion channels. Fabrication of dynamic, reversible and biocompatible scaffolds with non-invasive external triggers has so far been limited. Here, the authors report on the creation of 3D printed scaffolds with Janus structure that produce nanovibrations when exposed to ultrasound, promoting bone regeneration.
Collapse
|
40
|
Benya PD, Kavanaugh A, Zakarian M, Söderlind P, Jashashvili T, Zhang N, Waldorff EI, Ryaby JT, Billi F. Pulsed electromagnetic field (PEMF) transiently stimulates the rate of mineralization in a 3-dimensional ring culture model of osteogenesis. PLoS One 2021; 16:e0244223. [PMID: 33539401 PMCID: PMC7861434 DOI: 10.1371/journal.pone.0244223] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Accepted: 12/04/2020] [Indexed: 11/25/2022] Open
Abstract
Pulsed Electromagnetic Field (PEMF) has shown efficacy in bone repair and yet the optimum characteristics of this modality and its molecular mechanism remain unclear. To determine the effects of timing of PEMF treatment, we present a novel three-dimensional culture model of osteogenesis that demonstrates strong de novo generation of collagen and mineral matrix and exhibits stimulation by PEMF in multiple stages over 62 days of culture. Mouse postnatal day 2 calvarial pre-osteoblasts were cast within and around Teflon rings by polymerization of fibrinogen and cultured suspended without contact with tissue culture plastic. Ring constructs were exposed to PEMF for 4h/day for the entire culture (Daily), or just during Day1-Day10, Day11-Day 27, or Day28-Day63 and cultured without PEMF for the preceding or remaining days, and compared to no-PEMF controls. PEMF was conducted as HF Physio, 40.85 kHz frequency with a 67 ms burst period and an amplitude of 1.19 mT. Osteogenesis was kinetically monitored by repeated fluorescence measurements of continuously present Alizarin Red S (ARS) and periodically confirmed by micro-CT. PEMF treatment induced early-onset and statistically significant transient stimulation (~4-fold) of the mineralization rate when PEMF was applied Daily, or during D1-D10 and D11-D27. Stimulation was apparent but not significant between D28-D63 by ARS but was significant at D63 by micro-CT. PEMF also shifted the micro-CT density profiles to higher densities in each PEMF treatment group. Ring culture generated tissue with a mineral:matrix ratio of 2.0 by thermogravimetric analysis (80% of the calvaria control), and the deposited crystal structure was 50% hydroxyapatite by X-ray diffraction (63% of the calvaria and femur controls), independent of PEMF. These results were consistent with backscatter, secondary electron, and elemental analysis by scanning electron microscopy. Thus, in a defined, strong osteogenic environment, PEMF applied at different times was capable of further stimulation of osteogenesis with the potential to enhance bone repair.
Collapse
Affiliation(s)
- Paul D. Benya
- Department of Orthopaedic Surgery, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States of America
| | - Aaron Kavanaugh
- Department of Orthopaedic Surgery, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States of America
| | - Martin Zakarian
- Department of Orthopaedic Surgery, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States of America
| | - Philip Söderlind
- Department of Architecture and Urban Design, University of California Los Angeles, Los Angeles, California, United States of America
| | - Tea Jashashvili
- Department of Radiology, Molecular Imaging Center, Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
| | - Nianli Zhang
- Orthofix Medical Inc., Lewisville, Texas, United States of America
| | - Erik I. Waldorff
- Orthofix Medical Inc., Lewisville, Texas, United States of America
| | - James T. Ryaby
- Orthofix Medical Inc., Lewisville, Texas, United States of America
| | - Fabrizio Billi
- Department of Orthopaedic Surgery, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States of America
- * E-mail:
| |
Collapse
|
41
|
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: 10.8] [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
|
42
|
Gavazzo P, Viti F, Donnelly H, Oliva MAG, Salmeron-Sanchez M, Dalby MJ, Vassalli M. Biophysical phenotyping of mesenchymal stem cells along the osteogenic differentiation pathway. Cell Biol Toxicol 2021; 37:915-933. [PMID: 33420657 DOI: 10.1007/s10565-020-09569-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Accepted: 10/30/2020] [Indexed: 12/22/2022]
Abstract
Mesenchymal stem cells represent an important resource, for bone regenerative medicine and therapeutic applications. This review focuses on new advancements and biophysical tools which exploit different physical and chemical markers of mesenchymal stem cell populations, to finely characterize phenotype changes along their osteogenic differentiation process. Special attention is paid to recently developed label-free methods, which allow monitoring cell populations with minimal invasiveness. Among them, quantitative phase imaging, suitable for single-cell morphometric analysis, and nanoindentation, functional to cellular biomechanics investigation. Moreover, the pool of ion channels expressed in cells during differentiation is discussed, with particular interest for calcium homoeostasis.Altogether, a biophysical perspective of osteogenesis is proposed, offering a valuable tool for the assessment of the cell stage, but also suggesting potential physiological links between apparently independent phenomena.
Collapse
Affiliation(s)
- Paola Gavazzo
- Institute of Biophysics, National Research Council, Genoa, Italy
| | - Federica Viti
- Institute of Biophysics, National Research Council, Genoa, Italy.
| | - Hannah Donnelly
- Centre for the Cellular Microenvironment, Institute of Molecular, Cell and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Mariana Azevedo Gonzalez Oliva
- Centre for the Cellular Microenvironment, Division of Biomedical Engineering, School of Engineering, University of Glasgow, Glasgow, UK
| | - Manuel Salmeron-Sanchez
- Centre for the Cellular Microenvironment, Division of Biomedical Engineering, School of Engineering, University of Glasgow, Glasgow, UK
| | - Matthew J Dalby
- Centre for the Cellular Microenvironment, Institute of Molecular, Cell and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Massimo Vassalli
- Centre for the Cellular Microenvironment, Division of Biomedical Engineering, School of Engineering, University of Glasgow, Glasgow, UK
| |
Collapse
|
43
|
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
|
44
|
Zheng Z, Zhou T, Hu R, Huang M, Ao X, Chu J, Jiang T, Qin A, Zhang Z. A specific aggregation-induced emission-conjugated polymer enables visual monitoring of osteogenic differentiation. Bioact Mater 2020; 5:1018-1025. [PMID: 32695933 PMCID: PMC7355993 DOI: 10.1016/j.bioactmat.2020.06.020] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 06/10/2020] [Accepted: 06/28/2020] [Indexed: 01/10/2023] Open
Abstract
Osteogenic differentiation is the basis of bone growth and repair related to many diseases, in which evaluating the degree and ability of osteogenic transformation is quite important and highly desirable. However, fixing or stopping the growth of cells is required for conventional methods to monitor osteogenic differentiation, which cannot realize the full investigation of the dynamic process. Herein, a new anion conjugated polymer featuring aggregation-induced emission (AIE) characteristics is developed with excellent solubility for in-situ monitoring the process of osteogenic differentiation. This novel polymer can bind with osteogenic differentiated cells, and the intracellular fluorescence increases gradually with the enhancement of osteogenic differentiation. Moreover, it possesses good biosafety with negligible effect on cell activity and osteogenic differentiation, which cannot be realized by the typical method of Alizarin Red S staining. Further study shows that the polymer crosses the cell membrane through endocytosis and enriches in lysosomes, whereas no obvious fluorescence is detected with other cells, including non-differentiated osteoblast cells, under the same conditions, demonstrating the high selectivity. This is the first fluorescent probe with excellent specificity to realize real-time observation of the process of osteogenic differentiation. Therefore, PTB-EDTA shows great promise in the study of osteogenic differentiation and related applications.
Collapse
Affiliation(s)
- Zhenyu Zheng
- Department of Orthopaedics, The Third Affiliated Hospital, Southern Medical University, Academy of Orthopaedics, Guangdong Province, Guangzhou, 510630, China
| | - Taotao Zhou
- State Key Laboratory of Luminescent Materials and Devices, Guangdong Provincial Key Laboratory of Luminescence from Molecular Aggregates, Center for Aggregation-Induced Emission, South China University of Technology, Guangzhou, 510640, China
| | - Rong Hu
- State Key Laboratory of Luminescent Materials and Devices, Guangdong Provincial Key Laboratory of Luminescence from Molecular Aggregates, Center for Aggregation-Induced Emission, South China University of Technology, Guangzhou, 510640, China
| | - Minjun Huang
- Department of Orthopaedics, The Third Affiliated Hospital, Southern Medical University, Academy of Orthopaedics, Guangdong Province, Guangzhou, 510630, China
| | - Xiang Ao
- Department of Orthopaedics, The Third Affiliated Hospital, Southern Medical University, Academy of Orthopaedics, Guangdong Province, Guangzhou, 510630, China
| | - Jun Chu
- Department of Orthopaedics, The Third Affiliated Hospital, Southern Medical University, Academy of Orthopaedics, Guangdong Province, Guangzhou, 510630, China
| | - Tao Jiang
- Department of Orthopaedics, The Third Affiliated Hospital, Southern Medical University, Academy of Orthopaedics, Guangdong Province, Guangzhou, 510630, China
| | - Anjun Qin
- State Key Laboratory of Luminescent Materials and Devices, Guangdong Provincial Key Laboratory of Luminescence from Molecular Aggregates, Center for Aggregation-Induced Emission, South China University of Technology, Guangzhou, 510640, China
| | - Zhongmin Zhang
- Department of Orthopaedics, The Third Affiliated Hospital, Southern Medical University, Academy of Orthopaedics, Guangdong Province, Guangzhou, 510630, China
| |
Collapse
|
45
|
Dawson J, Lee PS, van Rienen U, Appali R. A General Theoretical Framework to Study the Influence of Electrical Fields on Mesenchymal Stem Cells. Front Bioeng Biotechnol 2020; 8:557447. [PMID: 33195123 PMCID: PMC7606877 DOI: 10.3389/fbioe.2020.557447] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 09/08/2020] [Indexed: 12/14/2022] Open
Abstract
Mesenchymal stem cell dynamics involve cell proliferation and cell differentiation into cells of distinct functional type, such as osteoblasts, adipocytes, or chondrocytes. Electrically active implants influence these dynamics for the regeneration of the cells in damaged tissues. How applied electric field influences processes of individual stem cells is a problem mostly unaddressed. The mathematical approaches to study stem cell dynamics have focused on the stem cell population as a whole, without resolving individual cells and intracellular processes. In this paper, we present a theoretical framework to describe the dynamics of a population of stem cells, taking into account the processes of the individual cells. We study the influence of the applied electric field on the cellular processes. We test our mean-field theory with the experiments from the literature, involving in vitro electrical stimulation of stem cells. We show that a simple model can quantitatively describe the experimentally observed time-course behavior of the total number of cells and the total alkaline phosphate activity in a population of mesenchymal stem cells. Our results show that the stem cell differentiation rate is dependent on the applied electrical field, confirming published experimental findings. Moreover, our analysis supports the cell density-dependent proliferation rate. Since the experimental results are averaged over many cells, our theoretical framework presents a robust and sensitive method for determining the effect of applied electric fields at the scale of the individual cell. These results indicate that the electric field stimulation may be effective in promoting bone regeneration by accelerating osteogenic differentiation.
Collapse
Affiliation(s)
- Jonathan Dawson
- Department of Computer Science and Electrical Engineering, Institute of General Electrical Engineering, University of Rostock, Rostock, Germany
| | - Poh Soo Lee
- Max Bergmann Center for Biomaterials, Institute for Materials Science, Technical University of Dresden, Dresden, Germany
| | - Ursula van Rienen
- Department of Computer Science and Electrical Engineering, Institute of General Electrical Engineering, University of Rostock, Rostock, Germany.,Department of Ageing of Individuals and Society, Interdisciplinary Faculty, University of Rostock, Rostock, Germany.,Department of Life, Light and Matter, Interdisciplinary Faculty, University of Rostock, Rostock, Germany
| | - Revathi Appali
- Department of Computer Science and Electrical Engineering, Institute of General Electrical Engineering, University of Rostock, Rostock, Germany.,Department of Ageing of Individuals and Society, Interdisciplinary Faculty, University of Rostock, Rostock, Germany
| |
Collapse
|
46
|
De Angelis A, Denzi A, Merla C, Andre FM, Mir LM, Apollonio F, Liberti M. Confocal Microscopy Improves 3D Microdosimetry Applied to Nanoporation Experiments Targeting Endoplasmic Reticulum. Front Bioeng Biotechnol 2020; 8:552261. [PMID: 33072718 PMCID: PMC7537786 DOI: 10.3389/fbioe.2020.552261] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Accepted: 08/24/2020] [Indexed: 12/12/2022] Open
Abstract
In the last years, microdosimetric numerical models of cells including intracellular compartments have been proposed, aiming to investigate the poration induced by the application of nanosecond pulsed electric fields (nsPEFs). A limitation of such models was the extremely approximate cell and organelle shapes, leading to an incorrect estimation of the electric field or transmembrane potential distribution in the studied domain. In order to obtain a reliable model of in vitro experiments and a one-to-one comparison between experimental and simulated results, here, a realistic model of 12 human mesenchymal stem cells was built starting from their optical microscopy images where different cell compartments were highlighted. The microdosimetric analysis of the cells group was quantified in terms of electric field and transmembrane potentials (TMPs) induced by an externally applied 10-ns trapezoidal pulse with rise and fall times of 2 ns, with amplitudes ranging from 2 to 30 MV/m. The obtained results showed that the plasma and endoplasmic reticulum (ER) membrane of each cell respond in a different way to the same electric field amplitude, depending on differences in shape, size, and position of the single cell with respect to the applied electric field direction. Therefore, also the threshold for an efficient electroporation is highly different from cell to cell. This difference was quantitatively estimated through the cumulative distribution function of the pore density for the plasma and ER membrane of each cell, representing the probability that a certain percentage of membrane has reached a specific value of pore density. By comparing the dose-response curves resulted from the simulations and those from the experimental study of De Menorval et al. (2016), we found a very good matching of results for plasma and ER membrane when 2% of the porated area is considered sufficient for permeabilizing the membrane. This result is worth of noting as it highlights the possibility to effectively predict the behavior of a cell (or of a population of cells) exposed to nsPEFs. Therefore, the microdosimetric realistic model described here could represent a valid tool in setting up more efficient and controlled electroporation protocols.
Collapse
Affiliation(s)
- Annalisa De Angelis
- Inter University Center for the Study of Electromagnetic Fields and Biological Systems (ICEmB) at Department of Electronic Engineering and Telecommunications (DIET), University of Rome "La Sapienza", Rome, Italy.,Center for Life Nano Science@Sapienza, Istituto Italiano di Tecnologia, Rome, Italy
| | - Agnese Denzi
- Inter University Center for the Study of Electromagnetic Fields and Biological Systems (ICEmB) at Department of Electronic Engineering and Telecommunications (DIET), University of Rome "La Sapienza", Rome, Italy
| | - Caterina Merla
- National Italian Agency for Energy, New Technologies and Sustainable Economic Development - Department of Sustainability (ENEA, SSPT) - Division of Health Protection Technologies, Rome, Italy
| | - Frank M Andre
- Université Paris-Saclay, Institut Gustave Roussy, CNRS, Metabolic and Systemic Aspects of Oncogenesis, Villejuif, France
| | - Lluis M Mir
- Université Paris-Saclay, Institut Gustave Roussy, CNRS, Metabolic and Systemic Aspects of Oncogenesis, Villejuif, France
| | - Francesca Apollonio
- Inter University Center for the Study of Electromagnetic Fields and Biological Systems (ICEmB) at Department of Electronic Engineering and Telecommunications (DIET), University of Rome "La Sapienza", Rome, Italy.,Center for Life Nano Science@Sapienza, Istituto Italiano di Tecnologia, Rome, Italy
| | - Micaela Liberti
- Inter University Center for the Study of Electromagnetic Fields and Biological Systems (ICEmB) at Department of Electronic Engineering and Telecommunications (DIET), University of Rome "La Sapienza", Rome, Italy.,Center for Life Nano Science@Sapienza, Istituto Italiano di Tecnologia, Rome, Italy
| |
Collapse
|
47
|
In Vitro Production of Calcified Bone Matrix onto Wool Keratin Scaffolds via Osteogenic Factors and Electromagnetic Stimulus. MATERIALS 2020; 13:ma13143052. [PMID: 32650489 PMCID: PMC7411850 DOI: 10.3390/ma13143052] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 07/03/2020] [Accepted: 07/04/2020] [Indexed: 12/15/2022]
Abstract
Pulsed electromagnetic field (PEMF) has drawn attention as a potential tool to improve the ability of bone biomaterials to integrate into the surrounding tissue. We investigated the effects of PEMF (frequency, 75 Hz; magnetic induction amplitude, 2 mT; pulse duration, 1.3 ms) on human osteoblast-like cells (SAOS-2) seeded onto wool keratin scaffolds in terms of proliferation, differentiation, and production of the calcified bone extracellular matrix. The wool keratin scaffold offered a 3D porous architecture for cell guesting and nutrient diffusion, suggesting its possible use as a filler to repair bone defects. Here, the combined approach of applying a daily PEMF exposure with additional osteogenic factors stimulated the cells to increase both the deposition of bone-related proteins and calcified matrix onto the wool keratin scaffolds. Also, the presence of SAOS-2 cells, or PEMF, or osteogenic factors did not influence the compression behavior or the resilience of keratin scaffolds in wet conditions. Besides, ageing tests revealed that wool keratin scaffolds were very stable and showed a lower degradation rate compared to commercial collagen sponges. It is for these reasons that this tissue engineering strategy, which improves the osteointegration properties of the wool keratin scaffold, may have a promising application for long term support of bone formation in vivo.
Collapse
|
48
|
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: 0.4] [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
|
49
|
Sr-HA scaffolds fabricated by SPS technology promote the repair of segmental bone defects. Tissue Cell 2020; 66:101386. [PMID: 32933709 DOI: 10.1016/j.tice.2020.101386] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 05/13/2020] [Accepted: 05/15/2020] [Indexed: 01/27/2023]
Abstract
BACKGROUND Ideal bone defect repair scaffolds should be biodegradable, biocompatible, bioactive, porous, and provide adequate mechanical support. However, it is challenging to fabricate such an ideal bone repair scaffold. Previously, we showed that 5 wt.% strontium-doped hydroxyapatite (Sr-HA) scaffolds prepared by spark plasma sintering (SPS) technology exhibited good biocompatibility. Moreover, unlike pure hydroxyapatite (HA) scaffolds, HA scaffolds containing strontium (Sr) exhibited superior bioactivity, higher proliferation rate of BMSCs and MG-63 osteoblast cells, as well as enhanced BMSCs differentiation. METHODS In this study, we prepared pure HA scaffolds and 5 wt.% strontium containing Sr-HA scaffolds by SPS technology without adhesive, ammonium bicarbonate as pore former. Subsequently, scanning electron microscope (SEM) and X-Ray diffraction (XRD) were used to characterize the properties of Sr-HA and HA scaffolds. The ability of the scaffolds to repair bone defects was evaluated using a critical-sized rabbit tibia-bone defect rabbit model. Thirty 3-month-old New Zealand white rabbits were randomly divided into three groups (blank control group, Sr-HA scaffolds implanted group and HA scaffolds implanted group) with 10 rabbits in each group. These rabbits are sacrificed after 8 weeks and 16 weeks of surgery, and the repair effects of each scaffold were evaluated with X-ray, micro-CT, and HE staining. The three-point bending test was employed to assess the mechanical property of repaired bones. RESULTS XRD pattern indicated that Sr-HA and HA scaffolds possess a similar crystal structure after sintering, and that incorporation of strontium did not form impure phase. SEM showed that the porosity of Sr-HA and HA scaffolds was about 40 %. Universal Testing Machine tests showed that Sr-HA scaffolds had better compressive strength than HA scaffolds. Bone defect was obvious, and the fibrous tissue was formed in the bone defects of rabbits in the blank control group after 8 weeks of surgery. Sr-HA and HA scaffolds enhanced osteointegration of the host bone, and extensive woven bone was formed on the surface of the Sr-HA scaffolds. After 16 weeks, the bone strump became blunt and a small amount of callus was formed in the blank control group. Comparatively, the scaffolds were substantially degraded in the Sr-HA scaffolds implanted group while scaffolds shadows still were observed in the HA implanted group. Bone remodeling and cavity recanalization were completely developed in the Sr-HA scaffolds group. The compressive strength of repaired bone in the Sr-HA scaffolds implantation group was higher than that of HA scaffolds implantation group after 8 weeks and 16 weeks of surgery. CONCLUSIONS Our results show that the Sr-HA composite scaffolds can effectively repair bone defects and have good biodegradable properties.
Collapse
|
50
|
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: 5.0] [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
|