1
|
Mathavan N, Singh A, Marques FC, Günther D, Kuhn GA, Wehrle E, Müller R. Spatial transcriptomics in bone mechanomics: Exploring the mechanoregulation of fracture healing in the era of spatial omics. SCIENCE ADVANCES 2025; 11:eadp8496. [PMID: 39742473 DOI: 10.1126/sciadv.adp8496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Accepted: 11/19/2024] [Indexed: 01/03/2025]
Abstract
In recent decades, the field of bone mechanobiology has sought experimental techniques to unravel the molecular mechanisms governing the phenomenon of mechanically regulated fracture healing. Each cell within a fracture site resides within different local microenvironments characterized by different levels of mechanical strain; thus, preserving the spatial location of each cell is critical in relating cellular responses to mechanical stimuli. Our spatial transcriptomics-based "mechanomics" platform facilitates spatially resolved analysis of the molecular profiles of cells with respect to their local in vivo mechanical environment by integrating time-lapsed in vivo micro-computed tomography, spatial transcriptomics, and micro-finite element analysis. We investigate the transcriptomic responses of cells as a function of the local strain magnitude by identifying the differential expression of genes in regions of high and low strain within a fracture site. Our platform thus has the potential to address fundamental open questions within the field and to discover mechano-responsive targets to enhance fracture healing.
Collapse
Affiliation(s)
| | - Amit Singh
- Institute for Biomechanics, ETH Zürich, Zürich, Switzerland
| | | | - Denise Günther
- Institute for Biomechanics, ETH Zürich, Zürich, Switzerland
| | - Gisela A Kuhn
- Institute for Biomechanics, ETH Zürich, Zürich, Switzerland
| | - Esther Wehrle
- Institute for Biomechanics, ETH Zürich, Zürich, Switzerland
- AO Research Institute Davos, Davos Platz, Switzerland
| | - Ralph Müller
- Institute for Biomechanics, ETH Zürich, Zürich, Switzerland
| |
Collapse
|
2
|
Attia HG, Hamouda MA, Alasmari S, El-Telbany DF, Alamri ZZ, Qahl SH, Alfaifi MY, Al-Sawahli MM, Abd El Wahed S. Polyvinyl Alcohol Capped Silver Nanostructures for Fortified Apoptotic Potential Against Human Laryngeal Carcinoma Cells Hep-2 Using Extremely-Low Frequency Electromagnetic Field. Int J Nanomedicine 2024; 19:9317-9332. [PMID: 39282575 PMCID: PMC11401528 DOI: 10.2147/ijn.s453689] [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: 01/23/2024] [Accepted: 06/22/2024] [Indexed: 09/19/2024] Open
Abstract
Purpose : Polyvinyl alcohol-capped silver nanostructures (cAgNSs) were investigated in order to enhance the cytotoxicity, pro-apoptotic, and oxidant patterns of in human laryngeal carcinoma Hep-2 cells by employing a 50 mT electromagnetic field (LEMF) for 30 min. Methods Wet chemical reduction was used to synthesize the cAgNSs, and after they had been capped with polyvinyl alcohol, they were specifically examined for particle size analysis and structural morphology. To visualize how the silver may attach to the protein targets, a molecular docking study was conducted. Estimation of cytotoxicity, cell cycle progression supported by mRNA expression of three apoptotic-promoting genes and one apoptotic-resisting. Results Particle size analysis results were a mean particle size of 157.3±0.5 nm, zeta potential value of -29.6 mV±1.5 mV, and polydispersity index of 0.31±0.05. Significantly reduction of IC50 against Hep-2 cells by around 6-fold was concluded. Also, we obtained suppression of the proliferation of Hep-2 cells, especially in the G0/G1 and S phases. Significant enhanced mRNA expression revealed enhanced induced CASP3, p53, and Beclin-1 mediated pro-apoptosis and induced NF-κB mediated autophagy in Hep-2 cells. Augmented levels of GR, ROS and MDA as oxidative stress biomarkers were also obtained. HE staining of Hep-2 cells exposed to cAgNSs and LEMF confirmed the enhanced apoptotic potential comparatively. Conclusion By conclusion, the developed nano-sized structures with the aid of extremely-low frequency electromagnetic field were successful to fortify the anti-cancer profile of cAgNSs in Hep-2 cells.
Collapse
Affiliation(s)
- Hany G Attia
- Department of Pharmacognosy, College of Pharmacy, Najran University, Najran, Saudi Arabia
| | - Mai Abdelhalim Hamouda
- Department of Oral Pathology, Faculty of Dentistry, King Salman International University, El-Tur, Egypt
| | - Saeed Alasmari
- Department of Biology, College of Science and Arts, Najran University, Najran 1988, Saudi Arabia
| | - Dalia F El-Telbany
- Department of Pharmaceutics, Faculty of Pharmacy, Modern University for Technology and Information (MTI), Cairo, Egypt
| | - Zaenah Zuhair Alamri
- Department of Biological Science, College of Science, University of Jeddah, Jeddah, Saudi Arabia
| | - Safa H Qahl
- Department of Biological Science, College of Science, University of Jeddah, Jeddah, Saudi Arabia
| | - Mohammad Y Alfaifi
- Department of Biology, College of Science, King Khalid University, Abha, Saudi Arabia
| | | | - Sara Abd El Wahed
- Department of Oral Pathology, Faculty of Dentistry, The British University in Egypt, Cairo, Egypt
| |
Collapse
|
3
|
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
|
4
|
Wang A, Ma X, Bian J, Jiao Z, Zhu Q, Wang P, Zhao Y. Signalling pathways underlying pulsed electromagnetic fields in bone repair. Front Bioeng Biotechnol 2024; 12:1333566. [PMID: 38328443 PMCID: PMC10847561 DOI: 10.3389/fbioe.2024.1333566] [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: 11/05/2023] [Accepted: 01/15/2024] [Indexed: 02/09/2024] Open
Abstract
Pulsed electromagnetic field (PEMF) stimulation is a prospective non-invasive and safe physical therapy strategy for accelerating bone repair. PEMFs can activate signalling pathways, modulate ion channels, and regulate the expression of bone-related genes to enhance osteoblast activity and promote the regeneration of neural and vascular tissues, thereby accelerating bone formation during bone repair. Although their mechanisms of action remain unclear, recent studies provide ample evidence of the effects of PEMF on bone repair. In this review, we present the progress of research exploring the effects of PEMF on bone repair and systematically elucidate the mechanisms involved in PEMF-induced bone repair. Additionally, the potential clinical significance of PEMF therapy in fracture healing is underscored. Thus, this review seeks to provide a sufficient theoretical basis for the application of PEMFs in bone repair.
Collapse
Affiliation(s)
- Aoao Wang
- Medical School of Chinese PLA, Beijing, China
| | - Xinbo Ma
- Department of Chemistry, Capital Normal University, Beijing, China
| | - Jiaqi Bian
- Senior Department of Orthopaedics, The Fourth Medical Center of PLA General Hospital, Beijing, China
| | | | - Qiuyi Zhu
- Medical School of Chinese PLA, Beijing, China
| | - Peng Wang
- Department of Neurosurgery, The First Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Yantao Zhao
- Senior Department of Orthopaedics, The Fourth Medical Center of PLA General Hospital, Beijing, China
| |
Collapse
|
5
|
Amirinejad M, Eftekhar-Vaghefi SH, Nematollahi Mahani SN, Salari M, Yahyapour R, Ahmadi-Zeidabadi M. Exposure to Low-Frequency Radiation Changes the Expression of Nestin, VEGF, BCRP and Apoptosis Markers During Glioma Treatment Strategy: An In Vitro Study. Curr Radiopharm 2024; 17:55-67. [PMID: 38817005 DOI: 10.2174/0118744710258350230921065159] [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: 04/17/2023] [Revised: 08/20/2023] [Accepted: 08/28/2023] [Indexed: 06/01/2024]
Abstract
BACKGROUND Exposure to physical contamination during chemotherapy, including non-ionizing electromagnetic fields, raises concerns about the widespread sources of exposure to this type of radiation. Glioblastoma multiforme (GBM) is an aggressive central nervous system tumor that is hard to treat due to resistance to drugs such as temozolomide (TMZ). OBJECTIVE Electromagnetic fields (EMF) and haloperidol (HLP) may have anticancer effects. In this study, we investigated the effects of TMZ, HLP, and EMF on GBM cell lines and analyzed the association between non-ionizing radiation and the risk of change in drug performance. METHODS Cell viability and reactive oxygen species (ROS) generation were measured by MTT and NBT assay, respectively. Then, the expression levels of breast cancer-resistant protein (BCRP), Bax, Bcl2, Nestin, vascular endothelial growth factor (VEGF) genes, and P53, Bax, and Bcl2 Proteins were evaluated by real-time PCR and western blot. RESULTS Co-treatment of GBM cells by HLP and TMZ enhanced apoptosis in T-98G and A172 cells by increasing the expression of P53 and Bax and decreasing Bcl-2. Interestingly, exposure of GBM cells to EMF decreased apoptosis in the TMZ+HLP group. CONCLUSION In conclusion, EMF reduced the synergistic effect of TMZ and HLP. This hypothesis that patients who are treated for brain tumors and suffer from depression should not be exposed to EMF is proposed in the present study. There appears to be an urgent need to reconsider exposure limits for low-frequency magnetic fields, based on experimental and epidemiological research, the relationship between exposure to non-ionizing radiation and adverse human health effects.
Collapse
Affiliation(s)
- Maryam Amirinejad
- Department of Anatomy, Afzalipour School of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Seyed Hassan Eftekhar-Vaghefi
- Department of Anatomy, Afzalipour School of Medicine, Kerman University of Medical Sciences, Kerman, Iran
- Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | | | | | - Rasoul Yahyapour
- School of Medicine, Jiroft University of Medical Sciences, Jiroft, Iran
| | - Meysam Ahmadi-Zeidabadi
- Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| |
Collapse
|
6
|
Franco-Obregón A. Harmonizing Magnetic Mitohormetic Regenerative Strategies: Developmental Implications of a Calcium-Mitochondrial Axis Invoked by Magnetic Field Exposure. Bioengineering (Basel) 2023; 10:1176. [PMID: 37892906 PMCID: PMC10604793 DOI: 10.3390/bioengineering10101176] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 10/05/2023] [Accepted: 10/08/2023] [Indexed: 10/29/2023] Open
Abstract
Mitohormesis is a process whereby mitochondrial stress responses, mediated by reactive oxygen species (ROS), act cumulatively to either instill survival adaptations (low ROS levels) or to produce cell damage (high ROS levels). The mitohormetic nature of extremely low-frequency electromagnetic field (ELF-EMF) exposure thus makes it susceptible to extraneous influences that also impinge on mitochondrial ROS production and contribute to the collective response. Consequently, magnetic stimulation paradigms are prone to experimental variability depending on diverse circumstances. The failure, or inability, to control for these factors has contributed to the existing discrepancies between published reports and in the interpretations made from the results generated therein. Confounding environmental factors include ambient magnetic fields, temperature, the mechanical environment, and the conventional use of aminoglycoside antibiotics. Biological factors include cell type and seeding density as well as the developmental, inflammatory, or senescence statuses of cells that depend on the prior handling of the experimental sample. Technological aspects include magnetic field directionality, uniformity, amplitude, and duration of exposure. All these factors will exhibit manifestations at the level of ROS production that will culminate as a unified cellular response in conjunction with magnetic exposure. Fortunately, many of these factors are under the control of the experimenter. This review will focus on delineating areas requiring technical and biological harmonization to assist in the designing of therapeutic strategies with more clearly defined and better predicted outcomes and to improve the mechanistic interpretation of the generated data, rather than on precise applications. This review will also explore the underlying mechanistic similarities between magnetic field exposure and other forms of biophysical stimuli, such as mechanical stimuli, that mutually induce elevations in intracellular calcium and ROS as a prerequisite for biological outcome. These forms of biophysical stimuli commonly invoke the activity of transient receptor potential cation channel classes, such as TRPC1.
Collapse
Affiliation(s)
- Alfredo Franco-Obregón
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore; ; Tel.: +65-6777-8427 or +65-6601-6143
- Institute of Health Technology and Innovation (iHealthtech), National University of Singapore, Singapore 117599, Singapore
- Biolonic Currents Electromagnetic Pulsing Systems Laboratory (BICEPS), National University of Singapore, Singapore 117599, Singapore
- NUS Centre for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
- Healthy Longevity Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117593, Singapore
- Nanomedicine Translational Research Programme, Centre for NanoMedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117544, Singapore
| |
Collapse
|
7
|
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
|
8
|
Linnemann C, Şahin F, Li N, Pscherer S, Götz F, Histing T, Nussler AK, Ehnert S. Insulin Can Delay Neutrophil Extracellular Trap Formation In Vitro-Implication for Diabetic Wound Care? BIOLOGY 2023; 12:1082. [PMID: 37626968 PMCID: PMC10452400 DOI: 10.3390/biology12081082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 07/24/2023] [Accepted: 07/31/2023] [Indexed: 08/27/2023]
Abstract
Diabetes is a worldwide evolving disease with many associated complications, one of which is delayed or impaired wound healing. Appropriate wound healing strongly relies on the inflammatory reaction directly after injury, which is often altered in diabetic wound healing. After an injury, neutrophils are the first cells to enter the wound site. They have a special defense mechanism, neutrophil extracellular traps (NETs), consisting of released DNA coated with antimicrobial proteins and histones. Despite being a powerful weapon against pathogens, NETs were shown to contribute to impaired wound healing in diabetic mice and are associated with amputations in diabetic foot ulcer patients. The anti-diabetic drugs metformin and liraglutide have already been shown to regulate NET formation. In this study, the effect of insulin was investigated. NET formation after stimulation with PMA (phorbol myristate acetate), LPS (lipopolysaccharide), or calcium ionophore (CI) in the presence/absence of insulin was analyzed. Insulin led to a robust delay of LPS- and PMA-induced NET formation but had no effect on CI-induced NET formation. Mechanistically, insulin induced reactive oxygen species, phosphorylated p38, and ERK, but reduced citrullination of histone H3. Instead, bacterial killing was induced. Insulin might therefore be a new tool for the regulation of NET formation during diabetic wound healing, either in a systemic or topical application.
Collapse
Affiliation(s)
- Caren Linnemann
- Siegfried Weller Institute for Trauma Research, BG Unfallklinik Tübingen, Eberhard Karls Universität Tuebingen, 72076 Tuebingen, Germany; (C.L.); (A.K.N.)
| | - Filiz Şahin
- Siegfried Weller Institute for Trauma Research, BG Unfallklinik Tübingen, Eberhard Karls Universität Tuebingen, 72076 Tuebingen, Germany; (C.L.); (A.K.N.)
| | - Ningna Li
- Microbial Genetics, Interfaculty Institute of Microbiology and Infection Medicine, Eberhard Karls Universität Tuebingen, 72076 Tuebingen, Germany
| | - Stefan Pscherer
- Siegfried Weller Institute for Trauma Research, BG Unfallklinik Tübingen, Eberhard Karls Universität Tuebingen, 72076 Tuebingen, Germany; (C.L.); (A.K.N.)
- Department of Internal Medicine III, Sophien- and Hufeland-Hospital, 99425 Weimar, Germany
| | - Friedrich Götz
- Microbial Genetics, Interfaculty Institute of Microbiology and Infection Medicine, Eberhard Karls Universität Tuebingen, 72076 Tuebingen, Germany
| | - Tina Histing
- Siegfried Weller Institute for Trauma Research, BG Unfallklinik Tübingen, Eberhard Karls Universität Tuebingen, 72076 Tuebingen, Germany; (C.L.); (A.K.N.)
| | - Andreas K. Nussler
- Siegfried Weller Institute for Trauma Research, BG Unfallklinik Tübingen, Eberhard Karls Universität Tuebingen, 72076 Tuebingen, Germany; (C.L.); (A.K.N.)
| | - Sabrina Ehnert
- Siegfried Weller Institute for Trauma Research, BG Unfallklinik Tübingen, Eberhard Karls Universität Tuebingen, 72076 Tuebingen, Germany; (C.L.); (A.K.N.)
| |
Collapse
|
9
|
Zhang C, Li H, Li J, Hu J, Yang K, Tao L. Oxidative stress: A common pathological state in a high-risk population for osteoporosis. Biomed Pharmacother 2023; 163:114834. [PMID: 37163779 DOI: 10.1016/j.biopha.2023.114834] [Citation(s) in RCA: 44] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 04/29/2023] [Accepted: 05/01/2023] [Indexed: 05/12/2023] Open
Abstract
Osteoporosis is becoming a major concern in the field of public health. The process of bone loss is insidious and does not directly induce obvious symptoms. Complications indicate an irreversible decrease in bone mass. The high-risk populations of osteoporosis, including postmenopausal women, elderly men, diabetic patients and obese individuals need regular bone mineral density testing and appropriate preventive treatment. However, the primary changes in these populations are different, increasing the difficulty of effective treatment of osteoporosis. Determining the core pathogenesis of osteoporosis helps improve the efficiency and efficacy of treatment among these populations. Oxidative stress is a common pathological state secondary to estrogen deficiency, aging, hyperglycemia and hyperlipemia. In this review, we divided oxidative stress into the direct effect of reactive oxygen species (ROS) and the reduction of antioxidant enzyme activity to discuss their roles in the development of osteoporosis. ROS initiated mitochondrial apoptotic signaling and suppressed osteogenic marker expression to weaken osteogenesis. MAPK and NF-κB signaling pathways mediated the positive effect of ROS on osteoclast differentiation. Antioxidant enzymes not only eliminate the negative effects of ROS, but also directly participate in the regulation of bone metabolism. Additionally, we also described the roles of proinflammatory factors and HIF-1α under the pathophysiological changes of inflammation and hypoxia, which provided a supplement of oxidative stress-induced osteoporosis. In conclusion, our review showed that oxidative stress was a common pathological state in a high-risk population for osteoporosis. Targeted oxidative stress treatment would greatly optimize the therapeutic schedule of various osteoporosis treatments.
Collapse
Affiliation(s)
- Chi Zhang
- Department of Orthopedics, First Hospital of China Medical University, No.155 Nanjing North Street, Shenyang, China
| | - Hao Li
- Department of Orthopedics, First Hospital of China Medical University, No.155 Nanjing North Street, Shenyang, China
| | - Jie Li
- Department of Orthopedics, First Hospital of China Medical University, No.155 Nanjing North Street, Shenyang, China
| | - Jiajin Hu
- Health Sciences Institute, China Medical University, Shenyang 110122, China
| | - Keda Yang
- Department of Orthopedics, First Hospital of China Medical University, No.155 Nanjing North Street, Shenyang, China.
| | - Lin Tao
- Department of Orthopedics, First Hospital of China Medical University, No.155 Nanjing North Street, Shenyang, China.
| |
Collapse
|
10
|
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:jfb14030165. [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
|
11
|
Zhao H, Liu C, Liu Y, Ding Q, Wang T, Li H, Wu H, Ma T. Harnessing electromagnetic fields to assist bone tissue engineering. Stem Cell Res Ther 2023; 14:7. [PMID: 36631880 PMCID: PMC9835389 DOI: 10.1186/s13287-022-03217-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Accepted: 12/08/2022] [Indexed: 01/13/2023] Open
Abstract
Bone tissue engineering (BTE) emerged as one of the exceptional means for bone defects owing to it providing mechanical supports to guide bone tissue regeneration. Great advances have been made to facilitate the success of BTE in regenerating bone within defects. The use of externally applied fields has been regarded as an alternative strategy for BTE. Electromagnetic fields (EMFs), known as a simple and non-invasive therapy, can remotely provide electric and magnetic stimulation to cells and biomaterials, thus applying EMFs to assist BTE would be a promising strategy for bone regeneration. When combined with BTE, EMFs improve cell adhesion to the material surface by promoting protein adsorption. Additionally, EMFs have positive effects on mesenchymal stem cells and show capabilities of pro-angiogenesis and macrophage polarization manipulation. These advantages of EMFs indicate that it is perfectly suitable for representing the adjuvant treatment of BTE. We also summarize studies concerning combinations of EMFs and diverse biomaterial types. The strategy of combining EMFs and BTE receives encouraging outcomes and holds a promising future for effectively treating bone defects.
Collapse
Affiliation(s)
- Hongqi Zhao
- grid.33199.310000 0004 0368 7223Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 Hubei China
| | - Chaoxu Liu
- grid.33199.310000 0004 0368 7223Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 Hubei China
| | - Yang Liu
- grid.33199.310000 0004 0368 7223Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 Hubei China
| | - Qing Ding
- grid.33199.310000 0004 0368 7223Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 Hubei China
| | - Tianqi Wang
- grid.33199.310000 0004 0368 7223Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 Hubei China
| | - Hao Li
- grid.33199.310000 0004 0368 7223Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 Hubei China
| | - Hua Wu
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China.
| | - Tian Ma
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China.
| |
Collapse
|
12
|
Redox Regulation of Autophagy in Cancer: Mechanism, Prevention and Therapy. LIFE (BASEL, SWITZERLAND) 2022; 13:life13010098. [PMID: 36676047 PMCID: PMC9863886 DOI: 10.3390/life13010098] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 12/18/2022] [Accepted: 12/22/2022] [Indexed: 12/31/2022]
Abstract
Reactive oxygen species (ROS), products of normal cellular metabolism, play an important role in signal transduction. Autophagy is an intracellular degradation process in response to various stress conditions, such as nutritional deprivation, organelle damage and accumulation of abnormal proteins. ROS and autophagy both exhibit double-edged sword roles in the occurrence and development of cancer. Studies have shown that oxidative stress, as the converging point of these stimuli, is involved in the mechanical regulation of autophagy process. The regulation of ROS on autophagy can be roughly divided into indirect and direct methods. The indirect regulation of autophagy by ROS includes post-transcriptional and transcriptional modulation. ROS-mediated post-transcriptional regulation of autophagy includes the post-translational modifications and protein interactions of AMPK, Beclin 1, PI3K and other molecules, while transcriptional regulation mainly focuses on p62/Keap1/Nrf2 pathway. Notably, ROS can directly oxidize key autophagy proteins, such as ATG4 and p62, leading to the inhibition of autophagy pathway. In this review, we will elaborate the molecular mechanisms of redox regulation of autophagy in cancer, and discuss ROS- and autophagy-based therapeutic strategies for cancer treatment.
Collapse
|
13
|
Aspera-Werz RH, Mück J, Linnemann C, Herbst M, Ihle C, Histing T, Nussler AK, Ehnert S. Nicotine and Cotinine Induce Neutrophil Extracellular Trap Formation-Potential Risk for Impaired Wound Healing in Smokers. Antioxidants (Basel) 2022; 11:antiox11122424. [PMID: 36552632 PMCID: PMC9774423 DOI: 10.3390/antiox11122424] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 11/28/2022] [Accepted: 12/06/2022] [Indexed: 12/13/2022] Open
Abstract
Smoking undoubtedly affects human health. Investigating 2318 representative patients at a level 1 trauma center identified delayed wound healing, tissue infections, and/or sepsis as main complications in smokers following trauma and orthopedic surgery. Therefore, smoking cessation is strongly advised to improve the clinical outcome in these patients, although smoking cessation often fails despite nicotine replacement therapy raising the need for specific interventions that may reduce the complication rate. However, the underlying mechanisms are still unknown. In diabetics, delayed wound healing and infections/sepsis are associated with increased neutrophilic PADI4 expression and formation of neutrophil extracellular traps (NETs). The aim was to investigate if similar mechanisms hold for smokers. Indeed, our results show higher PADI4 expression in active and heavy smokers than non-smokers, which is associated with an increased complication rate. However, in vitro stimulation of neutrophils with cigarette smoke extract (CSE) only moderately induced NET formation despite accumulation of reactive oxygen species (ROS). Physiological levels of nicotine and its main metabolite cotinine more effectively induced NET formation, although they did not actively induce the formation of ROS, but interfered with the activity of enzymes involved in anti-oxidative defense and NET formation. In summary, we propose increased formation of NETs as possible triggers for delayed wound healing, tissue infections, and/or sepsis in smokers after a major trauma and orthopedic surgery. Smoking cessation might reduce this effect. However, our data show that smoking cessation supported by nicotine replacement therapy should be carefully considered as nicotine and its metabolite cotinine effectively induced NET formation in vitro, even without active formation of ROS.
Collapse
|
14
|
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
|
15
|
Pharmacological inhibition of HDAC6 improves muscle phenotypes in dystrophin-deficient mice by downregulating TGF-β via Smad3 acetylation. Nat Commun 2022; 13:7108. [PMID: 36402791 PMCID: PMC9675748 DOI: 10.1038/s41467-022-34831-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 11/01/2022] [Indexed: 11/21/2022] Open
Abstract
The absence of dystrophin in Duchenne muscular dystrophy disrupts the dystrophin-associated glycoprotein complex resulting in skeletal muscle fiber fragility and atrophy, associated with fibrosis as well as microtubule and neuromuscular junction disorganization. The specific, non-conventional cytoplasmic histone deacetylase 6 (HDAC6) was recently shown to regulate acetylcholine receptor distribution and muscle atrophy. Here, we report that administration of the HDAC6 selective inhibitor tubastatin A to the Duchenne muscular dystrophy, mdx mouse model increases muscle strength, improves microtubule, neuromuscular junction, and dystrophin-associated glycoprotein complex organization, and reduces muscle atrophy and fibrosis. Interestingly, we found that the beneficial effects of HDAC6 inhibition involve the downregulation of transforming growth factor beta signaling. By increasing Smad3 acetylation in the cytoplasm, HDAC6 inhibition reduces Smad2/3 phosphorylation, nuclear translocation, and transcriptional activity. These findings provide in vivo evidence that Smad3 is a new target of HDAC6 and implicate HDAC6 as a potential therapeutic target in Duchenne muscular dystrophy.
Collapse
|
16
|
Ronniger M, Aguida B, Stacke C, Chen Y, Ehnert S, Erdmann N, Eschenburg G, Falldorf K, Pooam M, Wing A, Ahmad M. A Novel Method to Achieve Precision and Reproducibility in Exposure Parameters for Low-Frequency Pulsed Magnetic Fields in Human Cell Cultures. Bioengineering (Basel) 2022; 9:bioengineering9100595. [PMID: 36290562 PMCID: PMC9598188 DOI: 10.3390/bioengineering9100595] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 09/30/2022] [Accepted: 09/30/2022] [Indexed: 11/16/2022] Open
Abstract
The effects of extremely low-frequency electromagnetic field (ELF-MF) exposure on living systems have been widely studied at the fundamental level and also claimed as beneficial for the treatment of diseases for over 50 years. However, the underlying mechanisms and cellular targets of ELF-MF exposure remain poorly understood and the field has been plagued with controversy stemming from an endemic lack of reproducibility of published findings. To address this problem, we here demonstrate a technically simple and reproducible EMF exposure protocol to achieve a standardized experimental approach which can be readily adopted in any lab. As an assay system, we chose a commercially available inflammatory model human cell line; its response to magnetic fields involves changes in gene expression which can be monitored by a simple colorimetric reporter gene assay. The cells were seeded and cultured in microplates and inserted into a custom-built, semi-automated incubation and exposure system which accurately controls the incubation (temperature, humidity, CO2) and magnetic-field exposure conditions. A specific alternating magnetic field (<1.0% spatial variance) including far-field reduction provided defined exposure conditions at the position of each well of the microplate. To avoid artifacts, all environmental and magnetic-field exposure parameters were logged in real time throughout the duration of the experiment. Under these extensively controlled conditions, the effect of the magnetic field on the cell cultures as assayed by the standardized operating procedure was highly reproducible between experiments. As we could fully define the characteristics (frequency, intensity, duration) of the pulsed magnetic field signals at the position of the sample well, we were, for the first time, able to accurately determine the effect of changing single ELF-MF parameters such as signal shape, frequency, intensity and duty cycle on the biological response. One signal in particular (10 Hz, 50% duty cycle, rectangular, bipolar, 39.6μT) provided a significant reduction in cytokine reporter gene expression by 37% in our model cell culture line. In sum, the accuracy, environmental control and data-logging capacity of the semi-automated exposure system should greatly facilitate research into fundamental cellular response mechanisms and achieve the consistency necessary to bring ELF-MF/PEMF research results into the scientific mainstream.
Collapse
Affiliation(s)
- Michael Ronniger
- Sachtleben GmbH, 20251 Hamburg, Germany
- Correspondence: (M.R.); (M.A.); Tel.: +49-408-060-961-25 (M.R.); +33-014-427-2916 (M.A.)
| | - Blanche Aguida
- Photobiology Research Group, Sorbonne Université CNRS, 75005 Paris, France
| | | | - Yangmengfan Chen
- Department of Biology, Faculty of Science, Naresuan University, Phitsanulok 65000, Thailand
| | - Sabrina Ehnert
- Department of Biology, Faculty of Science, Naresuan University, Phitsanulok 65000, Thailand
| | | | | | | | - Marootpong Pooam
- Siegfried Weller Institute for Trauma Research, Department of Trauma and Reconstructive Surgery, BG Unfallklinik Tübingen, Eberhard Karls Universität Tübingen, 72076 Tübingen, Germany
| | | | - Margaret Ahmad
- Photobiology Research Group, Sorbonne Université CNRS, 75005 Paris, France
- Correspondence: (M.R.); (M.A.); Tel.: +49-408-060-961-25 (M.R.); +33-014-427-2916 (M.A.)
| |
Collapse
|
17
|
Zadeh-Haghighi H, Simon C. Magnetic field effects in biology from the perspective of the radical pair mechanism. J R Soc Interface 2022; 19:20220325. [PMID: 35919980 PMCID: PMC9346374 DOI: 10.1098/rsif.2022.0325] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Accepted: 07/14/2022] [Indexed: 04/07/2023] Open
Abstract
Hundreds of studies have found that weak magnetic fields can significantly influence various biological systems. However, the underlying mechanisms behind these phenomena remain elusive. Remarkably, the magnetic energies implicated in these effects are much smaller than thermal energies. Here, we review these observations, and we suggest an explanation based on the radical pair mechanism, which involves the quantum dynamics of the electron and nuclear spins of transient radical molecules. While the radical pair mechanism has been studied in detail in the context of avian magnetoreception, the studies reviewed here show that magnetosensitivity is widespread throughout biology. We review magnetic field effects on various physiological functions, discussing static, hypomagnetic and oscillating magnetic fields, as well as isotope effects. We then review the radical pair mechanism as a potential unifying model for the described magnetic field effects, and we discuss plausible candidate molecules for the radical pairs. We review recent studies proposing that the radical pair mechanism provides explanations for isotope effects in xenon anaesthesia and lithium treatment of hyperactivity, magnetic field effects on the circadian clock, and hypomagnetic field effects on neurogenesis and microtubule assembly. We conclude by discussing future lines of investigation in this exciting new area of quantum biology.
Collapse
Affiliation(s)
- Hadi Zadeh-Haghighi
- Department of Physics and Astronomy, University of Calgary, Calgary, Alberta, Canada T2N 1N4
- Institute for Quantum Science and Technology, University of Calgary, Calgary, Alberta, Canada T2N 1N4
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada T2N 1N4
| | - Christoph Simon
- Department of Physics and Astronomy, University of Calgary, Calgary, Alberta, Canada T2N 1N4
- Institute for Quantum Science and Technology, University of Calgary, Calgary, Alberta, Canada T2N 1N4
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada T2N 1N4
| |
Collapse
|
18
|
Reactive oxygen species, the trident of Neptune in the hands of hecate; role in different diseases, signaling pathways, and detection methods. Arch Biochem Biophys 2022; 728:109357. [DOI: 10.1016/j.abb.2022.109357] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 06/29/2022] [Accepted: 07/16/2022] [Indexed: 12/22/2022]
|
19
|
Giovale M, Novelli L, Persico L, Motta F, Rampoldi S, Galli R, Monteforte P, Doveri M, Bianchi G, Selmi C, Bottaro LC. Low-energy Pulsed Electromagnetic Field Therapy Reduces Pain in Fibromyalgia: A Randomized Single-blind Controlled Pilot Study. RHEUMATOLOGY AND IMMUNOLOGY RESEARCH 2022; 3:77-83. [PMID: 36465321 PMCID: PMC9524818 DOI: 10.2478/rir-2022-0013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Accepted: 03/05/2022] [Indexed: 06/17/2023]
Abstract
OBJECTIVES Fibromyalgia symptoms have a significant impact on the quality of life and respond poorly to medications. It has been hypothesized that the use of low-energy pulsed electromagnetic field (PEMF) induces neuroprotective effects that may interfere with pain perception. We explored the efficacy of PEMF in patients affected by fibromyalgia. METHODS Twenty-one females (median age 59 years, interquartile range [IQR] 16.5) affected by fibromyalgia were randomized to receive pulsed electromagnetic field-triple energy pain treatment (PEMF-TEPT) or placebo at T0 and at 4 weeks and 8 weeks. Fibromyalgia impact questionnaire (FIQ), widespread pain index (WPI), visual analog score (VAS) pain, symptom severity (SS) scale, and short form 36 (SF-36) health survey questionnaire have been evaluated. RESULTS Patients in the PEMF-TEPT group had a significantly higher reduction of WPI compared to placebo (mean difference -12.90 ± standard deviation [SD] 5.32 vs. -1.91 ± 4.55, difference in difference [DD] of -10.99; P < 0.001), of SS score (-4.10 ± 4.85 vs. -2.00 ± 2.32; DD = -2.1; P < 0.05), of VAS pain (-48 ± 30.75 vs. -16.82 ± 23.69; DD = -31.18; P < 0.01). They also reported a higher improvement of FIQ and SF-36, albeit not reaching statistical significance. CONCLUSION In our pilot controlled study, PEMF-TEPT appeared to be safe and improved fibromyalgia symptoms.
Collapse
Affiliation(s)
| | - Lucia Novelli
- Rheumatology and Clinical Immunology; IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Luca Persico
- Department of Economics, University of Genova, Genoa, Italy
| | - Francesca Motta
- Rheumatology and Clinical Immunology; IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
- Department of Biomedical Sciences, Humanitas University, Milan, Italy
| | | | - Rossana Galli
- Division of Rheumatology, ASL3 Genovese, Genoa, Italy
| | | | - Marica Doveri
- Division of Rheumatology, ASL3 Genovese, Genoa, Italy
| | | | - Carlo Selmi
- Rheumatology and Clinical Immunology; IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
- Department of Biomedical Sciences, Humanitas University, Milan, Italy
| | | |
Collapse
|
20
|
Panda AK, Sitaramgupta VSN, Pandya HJ, Basu B. Electrical waveform dependent osteogenesis on PVDF/BaTiO 3 composite using a customized and programmable cell stimulator. Biotechnol Bioeng 2022; 119:1578-1597. [PMID: 35244212 DOI: 10.1002/bit.28076] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 02/22/2022] [Accepted: 02/23/2022] [Indexed: 11/07/2022]
Abstract
Directing cellular functionalities using biomaterial-based bioelectronic stimulation remains a significant constraint in translating research outcomes to address specific clinical challenges. Electrical stimulation is now being clinically used as a therapeutic treatment option to promote bone tissue regeneration and to improve neuromuscular functionalities. However, the nature of the electrical waveforms during the stimulation and underlying biophysical rationale are still not scientifically well explored. Furthermore, bone-mimicking implant-based bioelectrical regulation of osteoinductivity has not been translated to clinics. The present study demonstrates the role of the waveform in electrical signal to direct differentiation of stem cells on an electroactive polymeric substrate, using monophasic DC, square wave, and biphasic wave. In this regard, an in-house electrical stimulation device has been fabricated for the uninterrupted delivery of programmed electrical signals to stem cells in culture. To provide a functional platform for stem cells to differentiate, barium titanate (BaTiO3 , BT) reinforced PVDF has been developed with mechanical properties similar to bone. The electrical stimulation of human mesenchymal stem cells (hMSCs) on PVDF/BT composite inhibited proliferation rate at day 7, indicating early commitment for differentiation. The phenotypical characteristics of DC stimulated hMSCs provided signatures of differentiation towards osteogenic lineage, which was subsequently confirmed using ALP assay, collagen deposition, matrix mineralization, and genetic expression. Our findings suggest that DC stimulation induced early osteogenesis in hMSCs with a higher level of intracellular reactive oxygen species (ROS), whereas the stimulation with square wave directed late osteogenesis with a lower ROS regeneration. In summary, the present study critically analyzes the role of electrical stimulation and its waveforms in regulating osteogenesis, without external biochemical differentiation inducers, on a bone-mimicking functional substrate. Such a strategy can potentially be adopted to develop orthopedic implant-based bioelectronic medicine for bone regeneration. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Asish Kumar Panda
- Laboratory for Biomaterials, Materials Research Centre, Indian Institute of Science, Bangalore, India
| | - V S N Sitaramgupta
- Department of Electronic Systems Engineering, Indian Institute of Science, Bangalore, India
| | - Hardik J Pandya
- Department of Electronic Systems Engineering, Indian Institute of Science, Bangalore, India
- Centre for Product Design and Manufacturing, Indian Institute of Science, Bangalore, India
| | - Bikramjit Basu
- Laboratory for Biomaterials, Materials Research Centre, Indian Institute of Science, Bangalore, India
- Centre for Biosystems Science and Engineering, Indian Institute of Science, Bangalore, India
| |
Collapse
|
21
|
Sołek P, Mytych J, Łannik E, Majchrowicz L, Koszła O, Koziorowska A, Koziorowski M. Cancer on-target: Selective enhancement of 3-bromopyruvate action by an electromagnetic field in vitro. Free Radic Biol Med 2022; 180:153-164. [PMID: 35063649 DOI: 10.1016/j.freeradbiomed.2022.01.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 12/25/2021] [Accepted: 01/13/2022] [Indexed: 11/18/2022]
Abstract
Cancer is one of the leading causes of death in the modern world. Nowadays, most often treatment methods used in clinical oncology are drug therapies applied as monotherapy or combined therapy. Additionally, recent studies focus on developing approaches with the use of a drug in combination with other factors, not only chemical, to improve the probability and magnitude of therapeutic responses and reduce the possibility of chemoresistance. Such a promising factor seems to be an electromagnetic field (EMF) application. Here, we tested the effect of continuous or pulsed EMF on human cancer cells of different origin treated or not with 3-bromopyruvate, a small and powerful alkylating agent with a broad spectrum of anticancer activities. We provide strong evidence suggesting that ELF-EMF potentiates the anti-cancer activity of 3BP in human cancer cells through inhibition of TNFα secretion leading to irreversible p21/p27-dependent G2/M cell cycle arrest and finally cancer cell death. Our findings suggest a novel approach combining pharmacotherapy with ELF-EMF. In conclusion, electromagnetic field seems to be a potential modulator of anti-cancer efficacy of 3BP while combined therapy offers off-target activity. These features contribute to the development of innovative therapeutic strategies for cancer treatment.
Collapse
Affiliation(s)
- Przemysław Sołek
- Department of Biopharmacy, Medical University of Lublin, 20-093, Lublin, Poland; Department of Biotechnology, Institute of Biology and Biotechnology, University of Rzeszow, 35-310, Rzeszow, Poland.
| | - Jennifer Mytych
- Department of Biotechnology, Institute of Biology and Biotechnology, University of Rzeszow, 35-310, Rzeszow, Poland
| | - Ewelina Łannik
- Department of Biotechnology, Institute of Biology and Biotechnology, University of Rzeszow, 35-310, Rzeszow, Poland
| | - Lena Majchrowicz
- BRAINCITY, Laboratory of Neurobiology, Nencki Institute of Experimental Biology PAS, 02-093, Warsaw, Poland
| | - Oliwia Koszła
- Department of Synthesis and Chemical Technology of Pharmaceutical Substances with Computer Modeling Laboratory, Faculty of Pharmacy, Medical University of Lublin, 20-093, Lublin, Poland
| | - Anna Koziorowska
- College of Natural Sciences, University of Rzeszow, 35-310, Rzeszow, Poland
| | - Marek Koziorowski
- Department of Biotechnology, Institute of Biology and Biotechnology, University of Rzeszow, 35-310, Rzeszow, Poland
| |
Collapse
|
22
|
Pooam M, Jourdan N, Aguida B, Dahon C, Baouz S, Terry C, Raad H, Ahmad M. Exposure to 1.8 GHz radiofrequency field modulates ROS in human HEK293 cells as a function of signal amplitude. Commun Integr Biol 2022; 15:54-66. [PMID: 35126804 PMCID: PMC8816398 DOI: 10.1080/19420889.2022.2027698] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
The modern telecommunications industry is ubiquitous throughout the world, with a significant percentage of the population using cellular phones on a daily basis. The possible physiological consequences of wireless emissions in the GHz range are therefore of major interest, but remain poorly understood. Here, we show that exposure to a 1.8 GHz carrier frequency in the amplitude range of household telecommunications induces the formation of ROS (Reactive Oxygen Species) in human HEK293 cultured cells. The ROS concentrations detected by fluorescent imaging techniques increased significantly after 15 minutes of RF field exposure, and were localized to both nuclear and cytosolic cellular compartments. qPCR analysis showed altered gene expression of both anti-oxidative (SOD, GPX, GPX, and CAT) and oxidative (Nox-2) enzymes. In addition, multiple genes previously identified as responsive to static magnetic fields were found to also be regulated by RF, suggesting common features in response mechanisms. By contrast, many RF effects showed evidence of hormesis, whereby biological responsivity does not occur linearly as a function of signal amplitude. Instead, biphasic dose response curves occur with ‘blind’ spots at certain signal amplitudes where no measureable response occurs. We conclude that modulation of intracellular ROS can be a direct consequence of RF exposure dependent on signal frequency and amplitude. Since changes in intracellular ROS may have both harmful and beneficial effects, these could provide the basis for many reported physiological effects of RF exposure.
Collapse
Affiliation(s)
- Marootpong Pooam
- Department of Biology, Faculty of Science, Naresuan University, Phitsanulok, Thailand
| | | | | | | | | | - Colin Terry
- Department of Biology, Xavier University, Cincinnati, Ohio, USA
| | - Haider Raad
- Department of Biology, Xavier University, Cincinnati, Ohio, USA
| | - Margaret Ahmad
- Sorbonne Université - CNRS, Paris, France.,Department of Biology, Xavier University, Cincinnati, Ohio, USA
| |
Collapse
|
23
|
Zadeh-Haghighi H, Simon C. Radical pairs can explain magnetic field and lithium effects on the circadian clock. Sci Rep 2022; 12:269. [PMID: 34997158 PMCID: PMC8742017 DOI: 10.1038/s41598-021-04334-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Accepted: 12/14/2021] [Indexed: 12/21/2022] Open
Abstract
Drosophila's circadian clock can be perturbed by magnetic fields, as well as by lithium administration. Cryptochromes are critical for the circadian clock. Further, the radical pairs in cryptochrome also can explain magnetoreception in animals. Based on a simple radical pair mechanism model of the animal magnetic compass, we show that both magnetic fields and lithium can influence the spin dynamics of the naturally occurring radical pairs and hence modulate the circadian clock's rhythms. Using a simple chemical oscillator model for the circadian clock, we show that the spin dynamics influence a rate in the chemical oscillator model, which translates into a change in the circadian period. Our model can reproduce the results of two independent experiments, magnetic field and lithium effects on the circadian clock. Our model predicts that stronger magnetic fields would shorten the clock's period. We also predict that lithium influences the clock in an isotope-dependent manner. Furthermore, our model also predicts that magnetic fields and hyperfine interactions modulate oxidative stress. The findings of this work suggest that the quantum nature of radical pairs might play roles in the brain, as another piece of evidence in addition to recent results on xenon anesthesia and lithium effects on hyperactivity.
Collapse
Affiliation(s)
- Hadi Zadeh-Haghighi
- Department of Physics and Astronomy, University of Calgary, Calgary, AB, T2N 1N4, Canada.
- Institute for Quantum Science and Technology, University of Calgary, Calgary, AB, T2N 1N4, Canada.
- Hotchkiss Brain Institute, University of Calgary, Calgary, AB, T2N 1N4, Canada.
| | - Christoph Simon
- Department of Physics and Astronomy, University of Calgary, Calgary, AB, T2N 1N4, Canada.
- Institute for Quantum Science and Technology, University of Calgary, Calgary, AB, T2N 1N4, Canada.
- Hotchkiss Brain Institute, University of Calgary, Calgary, AB, T2N 1N4, Canada.
| |
Collapse
|
24
|
Li Y, Yang Y, Wang M, Zhang X, Bai S, Lu X, Li Y, Waldorff EI, Zhang N, Lee WYW, Li G. High slew rate pulsed electromagnetic field enhances bone consolidation and shortens daily treatment duration in distraction osteogenesis. Bone Joint Res 2021; 10:767-779. [PMID: 34872332 PMCID: PMC8696558 DOI: 10.1302/2046-3758.1012.bjr-2021-0274.r1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Aims Distraction osteogenesis (DO) is a useful orthopaedic procedure employed to lengthen and reshape bones by stimulating bone formation through controlled slow stretching force. Despite its promising applications, difficulties are still encountered. Our previous study demonstrated that pulsed electromagnetic field (PEMF) treatment significantly enhances bone mineralization and neovascularization, suggesting its potential application. The current study compared a new, high slew rate (HSR) PEMF signal, with different treatment durations, with the standard Food and Drug Administration (FDA)-approved signal, to determine if HSR PEMF is a better alternative for bone formation augmentation. Methods The effects of a HSR PEMF signal with three daily treatment durations (0.5, one, and three hours/day) were investigated in an established rat DO model with comparison of an FDA-approved classic signal (three hrs/day). PEMF treatments were applied to the rats daily for 35 days, starting from the distraction phase until termination. Radiography, micro-CT (μCT), biomechanical tests, and histological examinations were employed to evaluate the quality of bone formation. Results All rats tolerated the treatment well and no obvious adverse effects were found. By comparison, the HSR signal (three hrs/day) treatment group achieved the best healing outcome, in that endochondral ossification and bone consolidation were enhanced. In addition, HSR signal treatment (one one hr/day) had similar effects to treatment using the classic signal (three three hrs/day), indicating that treatment duration could be significantly shortened with the HSR signal. Conclusion HSR signal may significantly enhance bone formation and shorten daily treatment duration in DO, making it a potential candidate for a new clinical protocol for patients undergoing DO treatments. Cite this article: Bone Joint Res 2021;10(12):767–779.
Collapse
Affiliation(s)
- Yucong Li
- Department of Orthopaedic and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China.,Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, China
| | - Yongkang Yang
- Department of Orthopaedic and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China.,Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, China
| | - Ming Wang
- Department of Orthopaedic and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China.,Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, China
| | - Xiaoting Zhang
- Department of Orthopaedic and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China.,Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, China
| | - Shanshan Bai
- Department of Orthopaedic and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China.,Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, China
| | - Xuan Lu
- Department of Orthopaedic and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China.,Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, China
| | - Yuan Li
- Department of Orthopaedic and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China.,Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, China
| | - Erik I Waldorff
- Research & Clinical Affairs, Orthofix Medical Inc, Lewisville, Texas, USA
| | - Nianli Zhang
- Research & Clinical Affairs, Orthofix Medical Inc, Lewisville, Texas, USA
| | - Wayne Yuk-Wai Lee
- Department of Orthopaedic and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China.,SH Ho Scoliosis Research Laboratory, Joint Scoliosis Research Center of the Chinese University of Hong Kong and Nanjing University, Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Hong Kong, China.,Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Gang Li
- Department of Orthopaedic and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China.,Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, China
| |
Collapse
|
25
|
Modulation of Macrophage Activity by Pulsed Electromagnetic Fields in the Context of Fracture Healing. Bioengineering (Basel) 2021; 8:bioengineering8110167. [PMID: 34821733 PMCID: PMC8615107 DOI: 10.3390/bioengineering8110167] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 10/22/2021] [Accepted: 10/27/2021] [Indexed: 01/05/2023] Open
Abstract
Delayed fracture healing and fracture non-unions impose an enormous burden on individuals and society. Successful healing requires tight communication between immune cells and bone cells. Macrophages can be found in all healing phases. Due to their high plasticity and long life span, they represent good target cells for modulation. In the past, extremely low frequency pulsed electromagnet fields (ELF-PEMFs) have been shown to exert cell-specific effects depending on the field conditions. Thus, the aim was to identify the specific ELF-PEMFs able to modulate macrophage activity to indirectly promote mesenchymal stem/stromal cell (SCP-1 cells) function. After a blinded screening of 22 different ELF-PEMF, two fields (termed A and B) were further characterized as they diversely affected macrophage function. These two fields have similar fundamental frequencies (51.8 Hz and 52.3 Hz) but are emitted in different groups of pulses or rather send-pause intervals. Macrophages exposed to field A showed a pro-inflammatory function, represented by increased levels of phospho-Stat1 and CD86, the accumulation of ROS, and increased secretion of pro-inflammatory cytokines. In contrast, macrophages exposed to field B showed anti-inflammatory and pro-healing functions, represented by increased levels of Arginase I, increased secretion of anti-inflammatory cytokines, and growth factors are known to induce healing processes. The conditioned medium from macrophages exposed to both ELF-PEMFs favored the migration of SCP-1 cells, but the effect was stronger for field B. Furthermore, the conditioned medium from macrophages exposed to field B, but not to field A, stimulated the expression of extracellular matrix genes in SCP-1 cells, i.e., COL1A1, FN1, and BGN. In summary, our data show that specific ELF-PEMFs may affect immune cell function. Thus, knowing the specific ELF-PEMFs conditions and the underlying mechanisms bears great potential as an adjuvant treatment to modulate immune responses during pathologies, e.g., fracture healing.
Collapse
|
26
|
Synthesis and Characterization of a Novel Biocompatible Alloy, Ti-Nb-Zr-Ta-Sn. Int J Mol Sci 2021; 22:ijms221910611. [PMID: 34638960 PMCID: PMC8509052 DOI: 10.3390/ijms221910611] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 09/24/2021] [Accepted: 09/27/2021] [Indexed: 12/14/2022] Open
Abstract
Many current-generation biomedical implants are fabricated from the Ti-6Al-4V alloy because it has many attractive properties, such as low density and biocompatibility. However, the elastic modulus of this alloy is much larger than that of the surrounding bone, leading to bone resorption and, eventually, implant failure. In the present study, we synthesized and performed a detailed analysis of a novel low elastic modulus Ti-based alloy (Ti-28Nb-5Zr-2Ta-2Sn (TNZTS alloy)) using a variety of methods, including scanning electron microscopy, transmission electron microscopy, X-ray diffraction, and tensile test. Additionally, the in vitro biocompatibility of the TNZTS alloy was evaluated using SCP-1, SaOs-2, and THP-1 cell lines and primary human osteoblasts. Compared to Ti-6Al-4V, the elastic modulus of TNZTS alloy was significantly lower, while measures of its in vitro biocompatibility are comparable. O2 plasma treatment of the surface of the alloy significantly increased its hydrophilicity and, hence, its in vitro biocompatibility. TNZTS alloy specimens did not induce the release of cytokines by macrophages, indicating that such scaffolds would not trigger inflammatory responses. The present results suggest that the TNZTS alloy may have potential as an alternative to Ti-6Al-4V.
Collapse
|
27
|
Hollenberg AM, Huber A, Smith CO, Eliseev RA. Electromagnetic stimulation increases mitochondrial function in osteogenic cells and promotes bone fracture repair. Sci Rep 2021; 11:19114. [PMID: 34580378 PMCID: PMC8476611 DOI: 10.1038/s41598-021-98625-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Accepted: 08/25/2021] [Indexed: 11/17/2022] Open
Abstract
Bone fracture is a growing public health burden and there is a clinical need for non-invasive therapies to aid in the fracture healing process. Previous studies have demonstrated the utility of electromagnetic (EM) fields in promoting bone repair; however, its underlying mechanism of action is unclear. Interestingly, there is a growing body of literature describing positive effects of an EM field on mitochondria. In our own work, we have previously demonstrated that differentiation of osteoprogenitors into osteoblasts involves activation of mitochondrial oxidative phosphorylation (OxPhos). Therefore, it was reasonable to propose that EM field therapy exerts bone anabolic effects via stimulation of mitochondrial OxPhos. In this study, we show that application of a low intensity constant EM field source on osteogenic cells in vitro resulted in increased mitochondrial membrane potential and respiratory complex I activity and induced osteogenic differentiation. In the presence of mitochondrial inhibitor antimycin A, the osteoinductive effect was reversed, confirming that this effect was mediated via increased OxPhos activity. Using a mouse tibial bone fracture model in vivo, we show that application of a low intensity constant EM field source enhanced fracture repair via improved biomechanical properties and increased callus bone mineralization. Overall, this study provides supporting evidence that EM field therapy promotes bone fracture repair through mitochondrial OxPhos activation.
Collapse
Affiliation(s)
- Alex M Hollenberg
- Center for Musculoskeletal Research, University of Rochester School of Medicine & Dentistry, Rochester, NY, USA
| | - Aric Huber
- Center for Musculoskeletal Research, University of Rochester School of Medicine & Dentistry, Rochester, NY, USA
| | - Charles O Smith
- Center for Musculoskeletal Research, University of Rochester School of Medicine & Dentistry, Rochester, NY, USA
| | - Roman A Eliseev
- Center for Musculoskeletal Research, University of Rochester School of Medicine & Dentistry, Rochester, NY, USA.
- University of Rochester Medical Center, 601 Elmwood Ave, Rm 1-8541, Rochester, NY, 14642, USA.
| |
Collapse
|
28
|
Anti-Oxidative and Immune Regulatory Responses of THP-1 and PBMC to Pulsed EMF Are Field-Strength Dependent. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2021; 18:ijerph18189519. [PMID: 34574442 PMCID: PMC8471206 DOI: 10.3390/ijerph18189519] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 09/07/2021] [Accepted: 09/08/2021] [Indexed: 12/26/2022]
Abstract
Innate immune cells react to electromagnetic fields (EMF) by generating reactive oxygen species (ROS), crucial intracellular messengers. Discrepancies in applied parameters of EMF studies, e.g., flux densities, complicate direct comparison of downstream anti-oxidative responses and immune regulatory signaling. We therefore compared the impact of different EMF flux densities in human leukemic THP1 cells and peripheral blood mononuclear cells (PBMC) of healthy donors to additionally consider a potential disparate receptivity based on medical origin. ROS levels increased in THP1 cells stimulated with lipopolysaccharide (LPS) after one hour of EMF exposure. Moreover, weak EMF mitigated the depletion of the reducing agent NAD(P)H in THP1. Neither of these effects occurred in PBMC. Landscaping transcriptional responses to varied EMF revealed elevation of the anti-oxidative enzymes PRDX6 (2-fold) and DHCR24 (6-fold) in THP1, implying involvement in lipid metabolism. Furthermore, our study confirmed anti-inflammatory effects of EMF by 6-fold increased expression of IL10. Strikingly, THP1 responded to weak EMF, while PBMC were primarily affected by strong EMF, yet with severe cellular stress and enhanced rates of apoptosis, indicated by HSP70 and caspase 3 (CASP3). Taken together, our results emphasize an altered susceptibility of immune cells of different origin and associate EMF-related effects with anti-inflammatory signaling and lipid metabolism.
Collapse
|
29
|
DNMT1 and miRNAs: possible epigenetics footprints in electromagnetic fields utilization in oncology. Med Oncol 2021; 38:125. [PMID: 34495398 DOI: 10.1007/s12032-021-01574-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Accepted: 09/01/2021] [Indexed: 10/20/2022]
Abstract
Many studies were performed to unravel the effects of different types of Electromagnetic fields (EMFs) on biological systems. Some studies were conducted to exploit EMFs for medical purposes mainly in cancer therapy. Although many studies suggest that the EMFs exposures can be effective in pre-clinical cancer issues, the treatment outcomes of these exposures on the cancer cells, especially at the molecular level, are challenging and overwhelmingly complicated yet. This article aims to review the epigenetic mechanisms that can be altered by EMFs exposures with the main emphasis on Extremely low frequency electromagnetic field (ELF-EMF). The epigenetic mechanisms are reversible and affected by environmental factors, thus, EMFs exposures can modulate these mechanisms. According to the reports, ELF-EMF exposures affect epigenetic machinery directly or through the molecular signaling pathways. ELF-EMF in association with DNA methylation, histone modification, miRNAs, and nucleosome remodeling could affect the homeostasis of cancer cells and play a role in DNA damage repairing, apoptosis induction, prevention of metastasis, differentiation, and cell cycle regulation. In general, the result of this study shows that ELF-EMF exposure probably can be effective in cancer epigenetic therapy, but more molecular and clinical investigations are needed to clarify the safe and specific dosimetric characteristics of ELF-EMF in practice.
Collapse
|
30
|
Chen Y, Aspera-Werz RH, Menger MM, Falldorf K, Ronniger M, Stacke C, Histing T, Nussler AK, Ehnert S. Exposure to 16 Hz Pulsed Electromagnetic Fields Protect the Structural Integrity of Primary Cilia and Associated TGF-β Signaling in Osteoprogenitor Cells Harmed by Cigarette Smoke. Int J Mol Sci 2021; 22:7036. [PMID: 34210094 PMCID: PMC8268780 DOI: 10.3390/ijms22137036] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 06/25/2021] [Accepted: 06/28/2021] [Indexed: 01/01/2023] Open
Abstract
Cigarette smoking (CS) is one of the main factors related to avoidable diseases and death across the world. Cigarette smoke consists of numerous toxic compounds that contribute to the development of osteoporosis and fracture nonunion. Exposure to pulsed electromagnetic fields (PEMF) was proven to be a safe and effective therapy to support bone fracture healing. The aims of this study were to investigate if extremely low frequency (ELF-) PEMFs may be beneficial to treat CS-related bone disease, and which effect the duration of the exposure has. In this study, immortalized human mesenchymal stem cells (SCP-1 cells) impaired by 5% cigarette smoke extract (CSE) were exposed to ELF-PEMFs (16 Hz) with daily exposure ranging from 7 min to 90 min. Cell viability, adhesion, and spreading were evaluated by Sulforhodamine B, Calcein-AM staining, and Phalloidin-TRITC/Hoechst 33342 staining. A migration assay kit was used to determine cell migration. Changes in TGF-β signaling were evaluated with an adenoviral Smad2/3 reporter assay, RT-PCR, and Western blot. The structure and distribution of primary cilia were analyzed with immunofluorescent staining. Our data indicate that 30 min daily exposure to a specific ELF-PEMF most effectively promoted cell viability, enhanced cell adhesion and spreading, accelerated migration, and protected TGF-β signaling from CSE-induced harm. In summary, the current results provide evidence that ELF-PEMF can be used to support early bone healing in patients who smoke.
Collapse
Affiliation(s)
- Yangmengfan Chen
- Siegfried Weller Research Institute at the BG Trauma Center Tübingen, Department of Trauma and Reconstructive Surgery, University of Tübingen, Schnarrenbergstr. 95, D-72076 Tübingen, Germany; (Y.C.); (R.H.A.-W.); (M.M.M.); (T.H.); (S.E.)
| | - Romina H. Aspera-Werz
- Siegfried Weller Research Institute at the BG Trauma Center Tübingen, Department of Trauma and Reconstructive Surgery, University of Tübingen, Schnarrenbergstr. 95, D-72076 Tübingen, Germany; (Y.C.); (R.H.A.-W.); (M.M.M.); (T.H.); (S.E.)
| | - Maximilian M. Menger
- Siegfried Weller Research Institute at the BG Trauma Center Tübingen, Department of Trauma and Reconstructive Surgery, University of Tübingen, Schnarrenbergstr. 95, D-72076 Tübingen, Germany; (Y.C.); (R.H.A.-W.); (M.M.M.); (T.H.); (S.E.)
| | - Karsten Falldorf
- Sachtleben GmbH, Haus Spectrum am UKE, Martinistraße 64, D-20251 Hamburg, Germany; (K.F.); (M.R.); (C.S.)
| | - Michael Ronniger
- Sachtleben GmbH, Haus Spectrum am UKE, Martinistraße 64, D-20251 Hamburg, Germany; (K.F.); (M.R.); (C.S.)
| | - Christina Stacke
- Sachtleben GmbH, Haus Spectrum am UKE, Martinistraße 64, D-20251 Hamburg, Germany; (K.F.); (M.R.); (C.S.)
| | - Tina Histing
- Siegfried Weller Research Institute at the BG Trauma Center Tübingen, Department of Trauma and Reconstructive Surgery, University of Tübingen, Schnarrenbergstr. 95, D-72076 Tübingen, Germany; (Y.C.); (R.H.A.-W.); (M.M.M.); (T.H.); (S.E.)
| | - Andreas K. Nussler
- Siegfried Weller Research Institute at the BG Trauma Center Tübingen, Department of Trauma and Reconstructive Surgery, University of Tübingen, Schnarrenbergstr. 95, D-72076 Tübingen, Germany; (Y.C.); (R.H.A.-W.); (M.M.M.); (T.H.); (S.E.)
| | - Sabrina Ehnert
- Siegfried Weller Research Institute at the BG Trauma Center Tübingen, Department of Trauma and Reconstructive Surgery, University of Tübingen, Schnarrenbergstr. 95, D-72076 Tübingen, Germany; (Y.C.); (R.H.A.-W.); (M.M.M.); (T.H.); (S.E.)
| |
Collapse
|
31
|
Özgün A, Garipcan B. Magnetic field-induced Ca 2+ intake by mesenchymal stem cells is mediated by intracellular Zn 2+ and accompanied by a Zn 2+ influx. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2021; 1868:119062. [PMID: 34033861 DOI: 10.1016/j.bbamcr.2021.119062] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 05/07/2021] [Accepted: 05/12/2021] [Indexed: 01/21/2023]
Abstract
Chronic exposure to magnetic fields (MFs) has a diverse range of effects on biological systems but definitive molecular mechanisms of the interaction remain largely unknown. One of the most frequently reported effects of MF exposure is an elevated concentration of intracellular Ca2+ through disputed pathways. Other prominent effects include increased oxidative stress and upregulation of neural markers through EGFR activation in stem cells. Further characterization of cascades triggered by MF exposure is hindered by the phenotype diversity of biological models used in the literature. In an attempt to reveal more mechanistic data in this field, we combined the most commonly used biological model and MF parameters with the most commonly reported effects of MFs. Based on clues from the pathways previously defined as sensitive to MFs (EGFR and Zn2+-binding enzymes), the roles of different types of channels (voltage gated Ca2+ channels, NMDA receptors, TRP channels) were inquired in the effects of 50 Hz MFs on bone marrow-derived mesenchymal stem cells. We report that, an influx of Zn2+ accompanies MF-induced Ca2+ intake, which is only attenuated by the broad-range inhibitor of TRP channels and store-operated Ca2+ entry (SOCE), 2-Aminoethoxydiphenyl borate (2-APB) among other blockers (memantine, nifedipine, ethosuximide and gabapentin). Interestingly, cation influx completely disappears when intracellular Zn2+ is chelated. Our results rule out voltage gated Ca2+ channels as a gateway to MF-induced Ca2+ intake and suggest Zn2+-related channels as a new focus in the field.
Collapse
Affiliation(s)
- Alp Özgün
- Institute of Biomedical Engineering, Boğaziçi University, Istanbul, Turkey
| | - Bora Garipcan
- Institute of Biomedical Engineering, Boğaziçi University, Istanbul, Turkey.
| |
Collapse
|
32
|
Oladnabi M, Mishan MA, Rezaeikanavi M, Zargari M, Sadeghi RN, Bagheri A. Correlation between ELF-PEMF exposure and Human RPE Cell Proliferation, Apoptosis and Gene Expression. J Ophthalmic Vis Res 2021; 16:202-211. [PMID: 34055258 PMCID: PMC8126745 DOI: 10.18502/jovr.v16i2.9084] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Accepted: 09/22/2020] [Indexed: 11/24/2022] Open
Abstract
Purpose Emerging evidence implies that electromagnetic fields (EMFs) can negatively affect angiogenesis. In this regard, the effects of extremely low frequency pulsed electromagnetic field (ELF-PEMF) exposure on the relative expression level of angiogenic factors involved in the pathogenesis of ocular disorders were evaluated in human retinal pigment epithelial (hRPE) cells in order to investigate a noninvasive therapeutic method for patients with several ocular diseases associated with neovascularization. Methods After separating hRPE cells from globes, hRPE cells were exposed to 15 mT of ELF-PEMF (120 Hz) at 5, 10, and 15 min for seven days. Cell proliferation and apoptosis of treated cells were evaluated via ELISA assay. Moreover, relative expression changes of HIF-1α, CTGF, VEGFA, MMP-2, cathepsin D, and E2F3 were performed using real-time RT-PCR. Results ELF-PEMF exposure had no significant effects on the apoptosis and proliferation rate of hRPE cells. Expression level of HIF-1α, CTGF, VEGFA, MMP-2, cathepsin D, and E2F3 was downregulated following 5 min of ELF-PEMF exposure. Conclusion As ELF-PEMF showed inhibitory effects on the expression of angiogenic genes in hRPE cells with no cytotoxic or proliferative side effects, it can be introduced as a useful procedure for managing angiogenesis induced by retinal pathogenesis, although more studies with adequate follow-up in animal models are needed.
Collapse
Affiliation(s)
- Morteza Oladnabi
- Stem Cell Research Center, Golestan University of Medical Sciences, Gorgan, Iran.,Ischemic Disorders Research Center, Golestan University of Medical Sciences, Gorgan, Iran.,Both authors contributed equally to the manuscript
| | - Mohammad Amir Mishan
- Ocular Tissue Engineering Research Center, Research Institute for Ophthalmology and Vision Science, Shahid Beheshti University of Medical Sciences, Tehran, Iran.,Both authors contributed equally to the manuscript
| | - Mozhgan Rezaeikanavi
- Ocular Tissue Engineering Research Center, Research Institute for Ophthalmology and Vision Science, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mehryar Zargari
- Department of Clinical Biochemistry and Medical Genetics, Molecular and Cell Biology Research Center, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Rouhallah Najjar Sadeghi
- Department of Clinical Biochemistry and Medical Genetics, Molecular and Cell Biology Research Center, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Abouzar Bagheri
- Department of Clinical Biochemistry and Medical Genetics, Molecular and Cell Biology Research Center, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| |
Collapse
|
33
|
Pooam M, Aguida B, Drahy S, Jourdan N, Ahmad M. Therapeutic application of light and electromagnetic fields to reduce hyper-inflammation triggered by COVID-19. Commun Integr Biol 2021; 14:66-77. [PMID: 33995820 PMCID: PMC8096326 DOI: 10.1080/19420889.2021.1911413] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 03/28/2021] [Accepted: 03/29/2021] [Indexed: 12/17/2022] Open
Abstract
COVID-19 - related morbidity is associated with exaggerated inflammation and cytokine production in the lungs, leading to acute respiratory failure. The cellular mechanisms underlying these so-called 'cytokine storms' are regulated through the Toll-like receptor 4 (TLR4) signaling pathway and by ROS (Reactive Oxygen Species). Both light (Photobiomodulation) and magnetic fields (e.g., Pulsed Electro Magnetic Field) stimulation are noninvasive therapies known to confer anti-inflammatory effects and regulate ROS signaling pathways. Here we show that daily exposure to two 10-minute intervals of moderate intensity infra-red light significantly lowered the inflammatory response induced via the TLR4 receptor signaling pathway in human cell cultures. Anti-inflammatory effects were likewise achieved by electromagnetic field exposure of cells to daily 10-minute intervals of either Pulsed Electromagnetic Fields (PEMF), or to Low-Level static magnetic fields. Because current illumination and electromagnetic field therapies have no known side effects, and are already approved for some medical uses, we have here developed protocols for verification in clinical trials of COVID-19 infection. These treatments are affordable, simple to implement, and may help to resolve the acute respiratory distress of COVID-19 patients both in the home and in the hospital.
Collapse
Affiliation(s)
- Marootpong Pooam
- Photobiology Research Group, Sorbonne Université - CNRS, Paris, France
- Department of Biology, Faculty of Science, Naresuan University, Phitsanulok, Thailand
| | - Blanche Aguida
- Photobiology Research Group, Sorbonne Université - CNRS, Paris, France
| | - Soria Drahy
- Photobiology Research Group, Sorbonne Université - CNRS, Paris, France
| | - Nathalie Jourdan
- Photobiology Research Group, Sorbonne Université - CNRS, Paris, France
| | - Margaret Ahmad
- Photobiology Research Group, Sorbonne Université - CNRS, Paris, France
- Xavier University, Cincinnati, Ohio, U.S.A
| |
Collapse
|
34
|
Zhu S, Aspera-Werz RH, Chen T, Weng W, Braun B, Histing T, Nüssler AK. Maqui berry extract prevents cigarette smoke induced oxidative stress in human osteoblasts in vitro. EXCLI JOURNAL 2021; 20:281-296. [PMID: 33628164 PMCID: PMC7898044 DOI: 10.17179/excli2020-3244] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Accepted: 02/04/2021] [Indexed: 12/16/2022]
Abstract
Oxidative stress which can be induced by cigarette smoke (CS) is associated with an altered osteoblast differentiation, and an inhibition of the mineralization process. Therefore, treatments focusing on reducing oxidative stress in osteoblasts could be a potential therapy supporting bone formation. Maqui berry extract (MBE) is the richest natural source of delphinidins with high antioxidant activity. In the present study, we pre-/ co-/ post-incubated MBE in cigarette smoke extract (CSE)-affected human osteoblasts (hOBs), to investigate the effects of MBE as an antioxidant on hOBs. Our results clearly showed that high concentrations of MBE are toxic for hOBs, while physiological concentrations of MBE have no negative effects in vitro. Physiological concentrations of MBE can reduce oxidative stress caused by CSE in hOBs by activating the antioxidative regulator Nrf2 and its regulated antioxidative enzymes. Moreover, the physiological concentration of MBE prevents the detrimental effects of CSE-induced oxidative damage on hOBs by increasing cell viability, differentiation capability and matrix mineralization. Pre-incubation with MBE showed a positive effect on the activation of the cellular antioxidant system in hOBs. Thus, we conclude that MBE at physiological concentrations can effectively protect osteoblasts from oxidative stress-induced damage by activating the cells' antioxidative defense system.
Collapse
Affiliation(s)
- Sheng Zhu
- Department of Traumatology, Eberhard Karls University Tübingen, BG Clinic, Siegfried Weller Institute, Schnarrenbergstraße 95, 72076 Tübingen, Germany
| | - Romina H Aspera-Werz
- Department of Traumatology, Eberhard Karls University Tübingen, BG Clinic, Siegfried Weller Institute, Schnarrenbergstraße 95, 72076 Tübingen, Germany
| | - Tao Chen
- Department of Traumatology, Eberhard Karls University Tübingen, BG Clinic, Siegfried Weller Institute, Schnarrenbergstraße 95, 72076 Tübingen, Germany
| | - Weidong Weng
- Department of Traumatology, Eberhard Karls University Tübingen, BG Clinic, Siegfried Weller Institute, Schnarrenbergstraße 95, 72076 Tübingen, Germany
| | - Bianca Braun
- Department of Traumatology, Eberhard Karls University Tübingen, BG Clinic, Siegfried Weller Institute, Schnarrenbergstraße 95, 72076 Tübingen, Germany
| | - Tina Histing
- Department of Traumatology, Eberhard Karls University Tübingen, BG Clinic, Siegfried Weller Institute, Schnarrenbergstraße 95, 72076 Tübingen, Germany
| | - Andreas K Nüssler
- Department of Traumatology, Eberhard Karls University Tübingen, BG Clinic, Siegfried Weller Institute, Schnarrenbergstraße 95, 72076 Tübingen, Germany
| |
Collapse
|
35
|
Klimek A, Rogalska J. Extremely Low-Frequency Magnetic Field as a Stress Factor-Really Detrimental?-Insight into Literature from the Last Decade. Brain Sci 2021; 11:174. [PMID: 33572550 PMCID: PMC7912337 DOI: 10.3390/brainsci11020174] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 01/23/2021] [Accepted: 01/27/2021] [Indexed: 12/13/2022] Open
Abstract
Biological effects of extremely low-frequency magnetic field (ELF-MF) and its consequences on human health have become the subject of important and recurrent public debate. ELF-MF evokes cell/organism responses that are characteristic to a general stress reaction, thus it can be regarded as a stress factor. Exposure to ELF-MF "turns on" different intracellular mechanisms into both directions: compensatory or deleterious ones. ELF-MF can provoke morphological and physiological changes in stress-related systems, mainly nervous, hormonal, and immunological ones. This review summarizes the ELF-MF-mediated changes at various levels of the organism organization. Special attention is placed on the review of literature from the last decade. Most studies on ELF-MF effects concentrate on its negative influence, e.g., impairment of behavior towards depressive and anxiety disorders; however, in the last decade there was an increase in the number of research studies showing stimulating impact of ELF-MF on neuroplasticity and neurorehabilitation. In the face of numerous studies on the ELF-MF action, it is necessary to systematize the knowledge for a better understanding of the phenomenon, in order to reduce the risk associated with the exposure to this factor and to recognize the possibility of using it as a therapeutic agent.
Collapse
Affiliation(s)
| | - Justyna Rogalska
- Department of Animal Physiology and Neurobiology, Faculty of Biological and Veterinary Sciences, Nicolaus Copernicus University, 87-100 Torun, Poland;
| |
Collapse
|
36
|
Emelianov VY, Preobrazhenskaia EV, Nikolaev NS. Evaluating the Effectiveness of Biophysical Methods of Osteogenesis Stimulation: Review. TRAUMATOLOGY AND ORTHOPEDICS OF RUSSIA 2021; 27:86-96. [DOI: https:/doi.org/10.21823/2311-2905-2021-27-1-86-96] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/14/2023]
Abstract
Background. Stimulation of osteogenesis (SO) by biophysical methods has been widely used in practice to accelerate healing or stimulate the healing of fractures with non-unions, since the middle of the XIX century. SO can be carried out by direct current electrostimulation, or indirectly by low-intensity pulsed ultrasound, capacitive electrical coupling stimulation, and pulsed electromagnetic field stimulation. SO simulates natural physiological processes: in the case of electrical stimulation, it changes the electromagnetic potential of damaged cell tissues in a manner similar to normal healing processes, or in the case of low-intensity pulsed ultrasound, it produces weak mechanical effects on the fracture area. SO increases the expression of factors and signaling pathways responsible for tissue regeneration and bone mineralization and ultimately accelerates bone union.The purpose of this review was to present the most up-to-date data from laboratory and clinical studies of the effectiveness of SO.Material and Methods. The results of laboratory studies and the final results of metaanalyses for each of the four SO methods published from 1959 to 2020 in the PubMed, EMBASE, and eLibrary databases are reviewed.Conclusion. The use of SO effectively stimulates the healing of fractures with the correct location of the sensors, compliance with the intensity and time of exposure, as well as the timing of use for certain types of fractures. In case of non-union or delayed union of fractures, spondylodesis, arthrodesis, preference should be given to non-invasive methods of SO. Invasive direct current stimulation can be useful for non-union of long bones, spondylodesis with the risk of developing pseudoarthrosis.
Collapse
|
37
|
Emelianov VY, Preobrazhenskaia EV, Nikolaev NS. Evaluating the Effectiveness of Biophysical Methods of Osteogenesis Stimulation: Review. TRAUMATOLOGY AND ORTHOPEDICS OF RUSSIA 2021; 27:86-96. [DOI: 10.21823/2311-2905-2021-27-1-86-96] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Background. Stimulation of osteogenesis (SO) by biophysical methods has been widely used in practice to accelerate healing or stimulate the healing of fractures with non-unions, since the middle of the XIX century. SO can be carried out by direct current electrostimulation, or indirectly by low-intensity pulsed ultrasound, capacitive electrical coupling stimulation, and pulsed electromagnetic field stimulation. SO simulates natural physiological processes: in the case of electrical stimulation, it changes the electromagnetic potential of damaged cell tissues in a manner similar to normal healing processes, or in the case of low-intensity pulsed ultrasound, it produces weak mechanical effects on the fracture area. SO increases the expression of factors and signaling pathways responsible for tissue regeneration and bone mineralization and ultimately accelerates bone union.The purpose of this review was to present the most up-to-date data from laboratory and clinical studies of the effectiveness of SO.Material and Methods. The results of laboratory studies and the final results of metaanalyses for each of the four SO methods published from 1959 to 2020 in the PubMed, EMBASE, and eLibrary databases are reviewed.Conclusion. The use of SO effectively stimulates the healing of fractures with the correct location of the sensors, compliance with the intensity and time of exposure, as well as the timing of use for certain types of fractures. In case of non-union or delayed union of fractures, spondylodesis, arthrodesis, preference should be given to non-invasive methods of SO. Invasive direct current stimulation can be useful for non-union of long bones, spondylodesis with the risk of developing pseudoarthrosis.
Collapse
|
38
|
Perez FP, Maloney B, Chopra N, Morisaki JJ, Lahiri DK. Repeated electromagnetic field stimulation lowers amyloid-β peptide levels in primary human mixed brain tissue cultures. Sci Rep 2021; 11:621. [PMID: 33436686 PMCID: PMC7804462 DOI: 10.1038/s41598-020-77808-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Accepted: 11/17/2020] [Indexed: 02/07/2023] Open
Abstract
Late Onset Alzheimer's Disease is the most common cause of dementia, characterized by extracellular deposition of plaques primarily of amyloid-β (Aβ) peptide and tangles primarily of hyperphosphorylated tau protein. We present data to suggest a noninvasive strategy to decrease potentially toxic Aβ levels, using repeated electromagnetic field stimulation (REMFS) in primary human brain (PHB) cultures. We examined effects of REMFS on Aβ levels (Aβ40 and Aβ42, that are 40 or 42 amino acid residues in length, respectively) in PHB cultures at different frequencies, powers, and specific absorption rates (SAR). PHB cultures at day in vitro 7 (DIV7) treated with 64 MHz, and 1 hour daily for 14 days (DIV 21) had significantly reduced levels of secreted Aβ40 (p = 001) and Aβ42 (p = 0.029) peptides, compared to untreated cultures. PHB cultures (DIV7) treated at 64 MHz, for 1 or 2 hour during 14 days also produced significantly lower Aβ levels. PHB cultures (DIV28) treated with 64 MHz 1 hour/day during 4 or 8 days produced a similar significant reduction in Aβ40 levels. 0.4 W/kg was the minimum SAR required to produce a biological effect. Exposure did not result in cellular toxicity nor significant changes in secreted Aβ precursor protein-α (sAPPα) levels, suggesting the decrease in Aβ did not likely result from redirection toward the α-secretase pathway. EMF frequency and power used in our work is utilized in human magnetic resonance imaging (MRI, thus suggesting REMFS can be further developed in clinical settings to modulate Aβ deposition.
Collapse
Affiliation(s)
- Felipe P Perez
- Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Medicine, Division of General Internal Medicine and Geriatrics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Bryan Maloney
- Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Psychiatry, Institute of Psychiatric Research, Neuroscience Research Center, Indiana University School of Medicine, 320 W. 15th St, Indianapolis, IN, 46201, USA
| | - Nipun Chopra
- Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Psychiatry, Institute of Psychiatric Research, Neuroscience Research Center, Indiana University School of Medicine, 320 W. 15th St, Indianapolis, IN, 46201, USA
| | - Jorge J Morisaki
- Department of Bioengineering, University of Illinois at Chicago, Chicago, IL, USA
| | - Debomoy K Lahiri
- Indiana University School of Medicine, Indianapolis, IN, USA.
- Department of Psychiatry, Institute of Psychiatric Research, Neuroscience Research Center, Indiana University School of Medicine, 320 W. 15th St, Indianapolis, IN, 46201, USA.
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA.
| |
Collapse
|
39
|
Pooam M, Jourdan N, El Esawi M, Sherrard RM, Ahmad M. HEK293 cell response to static magnetic fields via the radical pair mechanism may explain therapeutic effects of pulsed electromagnetic fields. PLoS One 2020; 15:e0243038. [PMID: 33270696 PMCID: PMC7714230 DOI: 10.1371/journal.pone.0243038] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Accepted: 11/15/2020] [Indexed: 01/01/2023] Open
Abstract
PEMF (Pulsed Electromagnetic Field) stimulation has been used for therapeutic purposes for over 50 years including in the treatment of memory loss, depression, alleviation of pain, bone and wound healing, and treatment of certain cancers. However, the underlying cellular mechanisms mediating these effects have remained poorly understood. In particular, because magnetic field pulses will induce electric currents in the stimulated tissue, it is unclear whether the observed effects are due to the magnetic or electric component of the stimulation. Recently, it has been shown that PEMFs stimulate the formation of ROS (reactive oxygen species) in human cell cultures by a mechanism that requires cryptochrome, a putative magnetosensor. Here we show by qPCR analysis of ROS-regulated gene expression that simply removing cell cultures from the Earth’s geomagnetic field by placing them in a Low-Level Field condition induces similar effects on ROS signaling as does exposure of cells to PEMF. This effect can be explained by the so-called Radical Pair mechanism, which provides a quantum physical means by which the rates and product yields (e.g. ROS) of biochemical redox reactions may be modulated by magnetic fields. Since transient cancelling of the Earth’s magnetic field can in principle be achieved by PEMF exposure, we propose that the therapeutic effects of PEMFs may be explained by the ensuing modulation of ROS synthesis. Our results could lead to significant improvements in the design and therapeutic applications of PEMF devices.
Collapse
Affiliation(s)
- Marootpong Pooam
- Sorbonne Université – CNRS, UMR8256 - IBPS, Paris, France
- Department of Biology, Faculty of Science, Naresuan University, Phitsanulok, Thailand
| | | | - Mohamed El Esawi
- Sorbonne Université – CNRS, UMR8256 - IBPS, Paris, France
- Botany Department, Faculty of Science, Tanta University, Tanta, Egypt
| | | | - Margaret Ahmad
- Sorbonne Université – CNRS, UMR8256 - IBPS, Paris, France
- Xavier University, Cincinnati, Ohio, United States of America
- * E-mail:
| |
Collapse
|
40
|
Li S, Wei C, Lv Y. Preparation and Application of Magnetic Responsive Materials in Bone Tissue Engineering. Curr Stem Cell Res Ther 2020; 15:428-440. [PMID: 31893995 DOI: 10.2174/1574888x15666200101122505] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 11/01/2019] [Accepted: 12/06/2019] [Indexed: 11/22/2022]
Abstract
At present, many kinds of materials are used for bone tissue engineering, such as polymer materials, metals, etc., which in general have good biocompatibility and mechanical properties. However, these materials cannot be controlled artificially after implantation, which may result in poor repair performance. The appearance of the magnetic response material enables the scaffolds to have the corresponding ability to the external magnetic field. Within the magnetic field, the magnetic response material can achieve the targeted release of the drug, improve the performance of the scaffold, and further have a positive impact on bone formation. This paper first reviewed the preparation methods of magnetic responsive materials such as magnetic nanoparticles, magnetic polymers, magnetic bioceramic materials and magnetic alloys in recent years, and then introduced its main applications in the field of bone tissue engineering, including promoting osteogenic differentiation, targets release, bioimaging, cell patterning, etc. Finally, the mechanism of magnetic response materials to promote bone regeneration was introduced. The combination of magnetic field treatment methods will bring significant progress to regenerative medicine and help to improve the treatment of bone defects and promote bone tissue repair.
Collapse
Affiliation(s)
- Song Li
- Mechanobiology and Regenerative Medicine Laboratory, Bioengineering College, Chongqing University, Chongqing, China
| | - Changling Wei
- Mechanobiology and Regenerative Medicine Laboratory, Bioengineering College, Chongqing University, Chongqing, China
| | - Yonggang Lv
- Mechanobiology and Regenerative Medicine Laboratory, Bioengineering College, Chongqing University, Chongqing, China
| |
Collapse
|
41
|
Guerra MF, Lacoste MG, Anzulovich AC, Makinistian L. Magnetic fields, cancer and circadian rhythms: hypotheses on the relevance of intermittence and cycling. Proc Biol Sci 2019; 286:20192337. [PMID: 31795871 DOI: 10.1098/rspb.2019.2337] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Affiliation(s)
- María Florencia Guerra
- Departamento de Física, Instituto de Física Aplicada (INFAP), Universidad Nacional de San Luis y Consejo Nacional de Investigaciones Científicas y Técnicas, Ejército de los Andes 950, 5700 San Luis, Argentina.,Laboratorio de Cronobiología, IMIBIO-SL (CONICET-UNSL), Ejército de los Andes 950, 5700 San Luis, Argentina
| | - María Gabriela Lacoste
- Laboratorio de Cronobiología, IMIBIO-SL (CONICET-UNSL), Ejército de los Andes 950, 5700 San Luis, Argentina.,Facultad de Química, Bioquímica y Farmacia, Universidad Nacional de San Luis, San Luis, Argentina
| | - Ana Cecilia Anzulovich
- Laboratorio de Cronobiología, IMIBIO-SL (CONICET-UNSL), Ejército de los Andes 950, 5700 San Luis, Argentina.,Facultad de Química, Bioquímica y Farmacia, Universidad Nacional de San Luis, San Luis, Argentina
| | - Leonardo Makinistian
- Departamento de Física, Instituto de Física Aplicada (INFAP), Universidad Nacional de San Luis y Consejo Nacional de Investigaciones Científicas y Técnicas, Ejército de los Andes 950, 5700 San Luis, Argentina
| |
Collapse
|
42
|
Ehnert S, Schröter S, Aspera-Werz RH, Eisler W, Falldorf K, Ronniger M, Nussler AK. Translational Insights into Extremely Low Frequency Pulsed Electromagnetic Fields (ELF-PEMFs) for Bone Regeneration after Trauma and Orthopedic Surgery. J Clin Med 2019; 8:jcm8122028. [PMID: 31756999 PMCID: PMC6947624 DOI: 10.3390/jcm8122028] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 11/15/2019] [Accepted: 11/18/2019] [Indexed: 02/07/2023] Open
Abstract
The finding that alterations in electrical potential play an important role in the mechanical stimulation of the bone provoked hype that noninvasive extremely low frequency pulsed electromagnetic fields (ELF-PEMF) can be used to support healing of bone and osteochondral defects. This resulted in the development of many ELF-PEMF devices for clinical use. Due to the resulting diversity of the ELF-PEMF characteristics regarding treatment regimen, and reported results, exposure to ELF-PEMFs is generally not among the guidelines to treat bone and osteochondral defects. Notwithstanding, here we show that there is strong evidence for ELF-PEMF treatment. We give a short, confined overview of in vitro studies investigating effects of ELF-PEMF treatment on bone cells, highlighting likely mechanisms. Subsequently, we summarize prospective and blinded studies, investigating the effect of ELF-PEMF treatment on acute bone fractures and bone fracture non-unions, osteotomies, spinal fusion, osteoporosis, and osteoarthritis. Although these studies favor the use of ELF-PEMF treatment, they likewise demonstrate the need for more defined and better controlled/monitored treatment modalities. However, to establish indication-oriented treatment regimen, profound knowledge of the underlying mechanisms in the sense of cellular pathways/events triggered is required, highlighting the need for more systematic studies to unravel optimal treatment conditions.
Collapse
Affiliation(s)
- Sabrina Ehnert
- Siegfried Weller Institute for Trauma Research, Depterment of Trauma and Reconstructive Surgery, BG Unfallklinik Tübingen, Eberhard Karls Universität Tübingen, D-72076 Tübingen, Germany; (S.S.); (R.H.A.-W.); (W.E.); (A.K.N.)
- Correspondence: or ; Tel.: +49-7071-606-1067
| | - Steffen Schröter
- Siegfried Weller Institute for Trauma Research, Depterment of Trauma and Reconstructive Surgery, BG Unfallklinik Tübingen, Eberhard Karls Universität Tübingen, D-72076 Tübingen, Germany; (S.S.); (R.H.A.-W.); (W.E.); (A.K.N.)
| | - Romina H. Aspera-Werz
- Siegfried Weller Institute for Trauma Research, Depterment of Trauma and Reconstructive Surgery, BG Unfallklinik Tübingen, Eberhard Karls Universität Tübingen, D-72076 Tübingen, Germany; (S.S.); (R.H.A.-W.); (W.E.); (A.K.N.)
| | - Wiebke Eisler
- Siegfried Weller Institute for Trauma Research, Depterment of Trauma and Reconstructive Surgery, BG Unfallklinik Tübingen, Eberhard Karls Universität Tübingen, D-72076 Tübingen, Germany; (S.S.); (R.H.A.-W.); (W.E.); (A.K.N.)
| | - Karsten Falldorf
- Sachtleben GmbH, Hamburg, Haus Spectrum am UKE, Martinistraße 64, D-20251 Hamburg, Germany; (K.F.); (M.R.)
| | - Michael Ronniger
- Sachtleben GmbH, Hamburg, Haus Spectrum am UKE, Martinistraße 64, D-20251 Hamburg, Germany; (K.F.); (M.R.)
| | - Andreas K. Nussler
- Siegfried Weller Institute for Trauma Research, Depterment of Trauma and Reconstructive Surgery, BG Unfallklinik Tübingen, Eberhard Karls Universität Tübingen, D-72076 Tübingen, Germany; (S.S.); (R.H.A.-W.); (W.E.); (A.K.N.)
| |
Collapse
|
43
|
Ziegler P, Nussler AK, Wilbrand B, Falldorf K, Springer F, Fentz AK, Eschenburg G, Ziegler A, Stöckle U, Maurer E, Ateschrang A, Schröter S, Ehnert S. Pulsed Electromagnetic Field Therapy Improves Osseous Consolidation after High Tibial Osteotomy in Elderly Patients-A Randomized, Placebo-Controlled, Double-Blind Trial. J Clin Med 2019; 8:jcm8112008. [PMID: 31744243 PMCID: PMC6912342 DOI: 10.3390/jcm8112008] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 11/13/2019] [Accepted: 11/15/2019] [Indexed: 11/25/2022] Open
Abstract
Extremely low-frequency pulsed electromagnetic field (ELF-PEMF) therapy is proposed to support bone healing after injuries and surgical procedures, being of special interest for elderly patients. This study aimed at investigating the effect of a specific ELF-PEMF, recently identified to support osteoblast function in vitro, on bone healing after high tibial osteotomy (HTO). Patients who underwent HTO were randomized to ELF-PEMF or placebo treatment, both applied by optically identical external devices 7 min per day for 30 days following surgery. Osseous consolidation was evaluated by post-surgical X-rays (7 and 14 weeks). Serum markers were quantified by ELISA. Data were compared by a two-sided t-test (α = 0.05). Device readouts showed excellent therapy compliance. Baseline parameters, including age, sex, body mass index, wedge height and blood cell count, were comparable between both groups. X-rays revealed faster osseous consolidation for ELF-PEMF compared to placebo treatment, which was significant in patients ≥50 years (∆mean = 0.68%/week; p = 0.003). Findings are supported by post-surgically increased bone-specific alkaline phosphatase serum levels following ELF-PEMF, compared to placebo (∆mean = 2.2 µg/L; p = 0.029) treatment. Adverse device effects were not reported. ELF-PEMF treatment showed a tendency to accelerate osseous consolidation after HTO. This effect was stronger and more significant for patients ≥50 years. This ELF-PEMF treatment might represent a promising adjunct to conventional therapy supporting osseous consolidation in elderly patients. Level of Evidence: I.
Collapse
Affiliation(s)
- Patrick Ziegler
- Siegfried Weller Institute for Trauma Research, Department of Trauma and Reconstructive Surgery, BG Unfallklinik Tübingen, Eberhard Karls Universität Tübingen, Schnarrenbergstr. 95, D-72076 Tübingen, Germany; (P.Z.); (B.W.); (E.M.); (A.A.); (S.S.); (S.E.)
| | - Andreas K. Nussler
- Siegfried Weller Institute for Trauma Research, Department of Trauma and Reconstructive Surgery, BG Unfallklinik Tübingen, Eberhard Karls Universität Tübingen, Schnarrenbergstr. 95, D-72076 Tübingen, Germany; (P.Z.); (B.W.); (E.M.); (A.A.); (S.S.); (S.E.)
- Correspondence: ; Tel.: +49-7071-606-1065
| | - Benjamin Wilbrand
- Siegfried Weller Institute for Trauma Research, Department of Trauma and Reconstructive Surgery, BG Unfallklinik Tübingen, Eberhard Karls Universität Tübingen, Schnarrenbergstr. 95, D-72076 Tübingen, Germany; (P.Z.); (B.W.); (E.M.); (A.A.); (S.S.); (S.E.)
| | - Karsten Falldorf
- Sachtleben GmbH, Haus Spectrum am UKE, Martinistraße 64, D-20251 Hamburg, Germany; (K.F.); (A.-K.F.); (G.E.)
| | - Fabian Springer
- Department of Diagnostic and Interventional Radiology, University of Tübingen, Hoppe-Seyler-Str. 3, D-72076 Tübingen, Germany;
| | - Anne-Kristin Fentz
- Sachtleben GmbH, Haus Spectrum am UKE, Martinistraße 64, D-20251 Hamburg, Germany; (K.F.); (A.-K.F.); (G.E.)
| | - Georg Eschenburg
- Sachtleben GmbH, Haus Spectrum am UKE, Martinistraße 64, D-20251 Hamburg, Germany; (K.F.); (A.-K.F.); (G.E.)
| | - Andreas Ziegler
- StatSol Lübeck, Moenring 2, D-23560 Lübeck, Germany;
- School of Mathematics, Statistics and Computer Science, University of KwaZulu-Natal, Pietermaritzburg, Scottsville 3209, South Africa
| | - Ulrich Stöckle
- Center for Musculoskeletal Surgery, Charité—University Medicine Berlin, Augustenburger Platz 1, D-13353 Berlin, Germany;
| | - Elke Maurer
- Siegfried Weller Institute for Trauma Research, Department of Trauma and Reconstructive Surgery, BG Unfallklinik Tübingen, Eberhard Karls Universität Tübingen, Schnarrenbergstr. 95, D-72076 Tübingen, Germany; (P.Z.); (B.W.); (E.M.); (A.A.); (S.S.); (S.E.)
| | - Atesch Ateschrang
- Siegfried Weller Institute for Trauma Research, Department of Trauma and Reconstructive Surgery, BG Unfallklinik Tübingen, Eberhard Karls Universität Tübingen, Schnarrenbergstr. 95, D-72076 Tübingen, Germany; (P.Z.); (B.W.); (E.M.); (A.A.); (S.S.); (S.E.)
| | - Steffen Schröter
- Siegfried Weller Institute for Trauma Research, Department of Trauma and Reconstructive Surgery, BG Unfallklinik Tübingen, Eberhard Karls Universität Tübingen, Schnarrenbergstr. 95, D-72076 Tübingen, Germany; (P.Z.); (B.W.); (E.M.); (A.A.); (S.S.); (S.E.)
| | - Sabrina Ehnert
- Siegfried Weller Institute for Trauma Research, Department of Trauma and Reconstructive Surgery, BG Unfallklinik Tübingen, Eberhard Karls Universität Tübingen, Schnarrenbergstr. 95, D-72076 Tübingen, Germany; (P.Z.); (B.W.); (E.M.); (A.A.); (S.S.); (S.E.)
| |
Collapse
|
44
|
Lai H. Exposure to Static and Extremely-Low Frequency Electromagnetic Fields and Cellular Free Radicals. Electromagn Biol Med 2019; 38:231-248. [PMID: 31450976 DOI: 10.1080/15368378.2019.1656645] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
This paper summarizes studies on changes in cellular free radical activities from exposure to static and extremely-low frequency (ELF) electromagnetic fields (EMF), particularly magnetic fields. Changes in free radical activities, including levels of cellular reactive oxygen (ROS)/nitrogen (RNS) species and endogenous antioxidant enzymes and compounds that maintain physiological free radical concentrations in cells, is one of the most consistent effects of EMF exposure. These changes have been reported to affect many physiological functions such as DNA damage; immune response; inflammatory response; cell proliferation and differentiation; wound healing; neural electrical activities; and behavior. An important consideration is the effects of EMF-induced changes in free radicals on cell proliferation and differentiation. These cellular processes could affect cancer development and proper growth and development in organisms. On the other hand, they could cause selective killing of cancer cells, for instance, via the generation of the highly cytotoxic hydroxyl free radical by the Fenton Reaction. This provides a possibility of using these electromagnetic fields as a non-invasive and low side-effect cancer therapy. Static- and ELF-EMF probably play important roles in the evolution of living organisms. They are cues used in many critical survival functions, such as foraging, migration, and reproduction. Living organisms can detect and respond immediately to low environmental levels of these fields. Free radical processes are involved in some of these mechanisms. At this time, there is no credible hypothesis or mechanism that can adequately explain all the observed effects of static- and ELF-EMF on free radical processes. We are actually at the impasse that there are more questions than answers.
Collapse
Affiliation(s)
- Henry Lai
- Department of Bioengineering, University of Washington , Seattle , WA , USA
| |
Collapse
|
45
|
Galli C, Colangelo M, Pedrazzi G, Guizzardi S. The Response of Osteoblasts and Bone to Sinusoidal Electromagnetic Fields: Insights from the Literature. Calcif Tissue Int 2019; 105:127-147. [PMID: 30997574 DOI: 10.1007/s00223-019-00554-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Accepted: 04/11/2019] [Indexed: 12/23/2022]
Abstract
Electromagnetic fields (EMFs) have been proposed as a tool to ameliorate bone formation and healing. Despite their promising results, however, they have failed to enter routine clinical protocols to treat bone conditions where higher bone mass has to be achieved. This is no doubt also due to a fundamental lack of knowledge and understanding on their effects and the optimal settings for attaining the desired therapeutic effects. This review analysed the available in vitro and in vivo studies that assessed the effects of sinusoidal EMFs (SEMFs) on bone and bone cells, comparing the results and investigating possible mechanisms of action by which SEMFs interact with tissues and cells. The effects of SEMFs on bone have not been as thoroughly investigated as pulsed EMFs; however, abundant evidence shows that SEMFs affect the proliferation and differentiation of osteoblastic cells, acting on multiple cellular mechanisms. SEMFs have also proven to increase bone mass in rodents under normal conditions and in osteoporotic animals.
Collapse
Affiliation(s)
- C Galli
- Department of Medicine and Surgery, University of Parma, Parma, Italy.
| | - M Colangelo
- Department of Medicine and Surgery, Histology and Embryology Lab, University of Parma, Parma, Italy
| | - G Pedrazzi
- Department of Medicine and Surgery, Neuroscience Unit, University of Parma, Via Volturno 39, 43126, Parma, Italy
| | - S Guizzardi
- Department of Medicine and Surgery, Histology and Embryology Lab, University of Parma, Parma, Italy
| |
Collapse
|
46
|
Ahmadi-Zeidabadi M, Akbarnejad Z, Esmaeeli M, Masoumi-Ardakani Y, Mohammadipoor-Ghasemabad L, Eskandary H. Impact of extremely low-frequency electromagnetic field (100 Hz, 100 G) exposure on human glioblastoma U87 cells during Temozolomide administration. Electromagn Biol Med 2019; 38:198-209. [PMID: 31179753 DOI: 10.1080/15368378.2019.1625784] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Glioblastoma multiforme (GBM) is a highly malignant brain tumor with an extremely dismal prognosis, a median survival is12 months. Temozolomide (TMZ) is an alkylating agent widely used to treat cancer, resistance to this drug is often found. One unexplored possibility for overcoming this resistance is a treatment based on concomitant exposure to electromagnetic fields (EMF) and TMZ. Indeed, many evidences show that EMF affects cancer cells and drug performance. Therefore, the present study was carried out to evaluate the potential synergistic effect of 100 µM TMZ and EMF (100 Hz, 100 G) on human glioma cell line U87 U87 cells with four experimental groups (I-IV) were exposed to ELF-EMF and TMZ for 120 and 144 h, as follows: (I) control; (II) ELF-EMF; (III) TMZ; (IV) ELF-PEMFs / TMZ. mRNA expression of genes such as (Nestin,CD133, Notch4 and GFAP) were investigated by Real-time PCR and western blot. We also evaluated, SOD activity, MDA and calcium concentration by ELISA assay. Co-treatment synergistically decreased the expression of Nestin,CD133, and Notch4 and increased the GFAP genes. We also observed an increase in Superoxide dismutase (SOD) activity, Malondialdehyde (MDA) and Ca2+concentration in comparison to controls.TMZ prevents cancer progression not only through the induction of cell death, but also by inducing differentiation in cancer cells. In addition, our data demonstrate ELF-EMF (100 Hz, 100 G) can significantly enhance the effects of TMZ on human glioblastoma U87 cell. These findings may open new window for future studies.
Collapse
Affiliation(s)
- Meysam Ahmadi-Zeidabadi
- a Neuroscience Research Center, Institute of Neuropharmacology , Kerman University of Medical Sciences , Kerman , Iran
| | - Zeinab Akbarnejad
- b ENT and Head & Neck Research center and department, Hazrat Rasoul Hospital , The five senses Institute, Iran University of medical sciences , Tehran , Iran
| | - Marzie Esmaeeli
- a Neuroscience Research Center, Institute of Neuropharmacology , Kerman University of Medical Sciences , Kerman , Iran
| | - Yaser Masoumi-Ardakani
- c Physiology Research Center, Institute of Basic and Clinical Physiology Science , Kerman University of Medical Sciences , Kerman , Iran
| | | | - Hossein Eskandary
- a Neuroscience Research Center, Institute of Neuropharmacology , Kerman University of Medical Sciences , Kerman , Iran.,e Afzal Research Institute (NGO) , Kerman , Iran
| |
Collapse
|
47
|
Oltean-Dan D, Dogaru GB, Apostu D, Mester A, Benea HRC, Paiusan MG, Popa CO, Jianu EM, Bodizs GI, Berce C, Toader AM, Tomoaia G. Enhancement of bone consolidation using high-frequency pulsed electromagnetic fields (HF-PEMFs): An experimental study on rats. Bosn J Basic Med Sci 2019; 19:201-209. [PMID: 30794499 DOI: 10.17305/bjbms.2019.3854] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Accepted: 10/13/2018] [Indexed: 11/16/2022] Open
Abstract
In vitro studies showed that high-frequency pulsed electromagnetic fields (HF-PEMFs) increase the activity/expression of early and late osteogenic markers and enhance bone mineralization. The main aim of this study was to investigate the in vivo effects of HF-PEMFs on fracture healing using a rat model. A femur fracture was established by surgery in 20 male Wistar rats. Titanium nails were implanted to reduce and stabilize the fracture. After surgery, 20 rats were equally divided into untreated control and treated group (from the first postoperative day HF-PEMFs at 400 pulses/sec [pps] were applied for 10 minutes/day, for two weeks). Quantitative and qualitative assessment of bone formation was made at two and eight weeks following surgery and included morphological and histological analysis, serological analysis by ELISA, micro-computed tomography (micro-CT), and three-point bending test. At two weeks in HF-PEMF group, soft callus was at a more advanced fibrocartilaginous stage and the bone volume/total tissue volume (BV/TV) ratio in the callus area was significantly higher compared to control group (p = 0.047). Serum concentration of alkaline phosphatase (ALP) and osteocalcin (OC) was significantly higher in HF-PEMF group (ALP p = 0.026, OC p = 0.006) as well as the mechanical strength of femurs (p = 0.03). At eight weeks, femurs from HF-PEMF group had a completely formed woven bone with dense trabeculae, active bone marrow, and had a significantly higher BV/TV ratio compared to control (p = 0.01). HF-PEMFs applied from the first postoperative day, 10 minutes/day for two weeks, enhance bone consolidation in rats, especially in the early phase of fracture healing.
Collapse
Affiliation(s)
- Daniel Oltean-Dan
- "Iuliu Hatieganu" University of Medicine and Pharmacy, Cluj-Napoca, Romania.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Tuhanioğlu B, Erkan SO, Gürgen SG, Özdaş T, Görgülü O, Çiçek F, Günay İ. The effect of very low dose pulsed magnetic waves on cochlea. Braz J Otorhinolaryngol 2019; 85:282-289. [PMID: 30583943 PMCID: PMC9442804 DOI: 10.1016/j.bjorl.2018.10.013] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Revised: 10/05/2018] [Accepted: 10/28/2018] [Indexed: 01/15/2023] Open
Affiliation(s)
- Birgül Tuhanioğlu
- Health Science University Adana City Hospital, Department of Otorhinolaryngology-Head and Neck Surgery, Adana, Turkey.
| | - Sanem Okşan Erkan
- Health Science University Adana City Hospital, Department of Otorhinolaryngology-Head and Neck Surgery, Adana, Turkey
| | - Seren Gülşen Gürgen
- Celal Bayar University, Department of Histology and Embriology, Manisa, Turkey
| | - Talih Özdaş
- Health Science University Adana City Hospital, Department of Otorhinolaryngology-Head and Neck Surgery, Adana, Turkey
| | - Orhan Görgülü
- Health Science University Adana City Hospital, Department of Otorhinolaryngology-Head and Neck Surgery, Adana, Turkey
| | - Figen Çiçek
- Çukurova University, Department of Biophysics, Adana, Turkey
| | - İsmail Günay
- Çukurova University, Department of Biophysics, Adana, Turkey
| |
Collapse
|
49
|
Galli C, Pedrazzi G, Guizzardi S. The cellular effects of Pulsed Electromagnetic Fields on osteoblasts: A review. Bioelectromagnetics 2019; 40:211-233. [PMID: 30908726 DOI: 10.1002/bem.22187] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Accepted: 03/08/2019] [Indexed: 12/12/2022]
Abstract
Electromagnetic fields (EMFs) have long been known to interact with living organisms and their cells and to bear the potential for therapeutic use. Among the most extensively investigated applications, the use of Pulsed EMFs (PEMFs) has proven effective to ameliorate bone healing in several studies, although the evidence is still inconclusive. This is due in part to our still-poor understanding of the mechanisms by which PEMFs act on cells and affect their functions and to an ongoing lack of consensus on the most effective parameters for specific clinical applications. The present review has compared in vitro studies on PEMFs on different osteoblast models, which elucidate potential mechanisms of action for PEMFs, up to the most recent insights into the role of primary cilia, and highlight the critical issues underlying at least some of the inconsistent results in the available literature. Bioelectromagnetics. 2019;9999:XX-XX. © 2019 Bioelectromagnetics Society.
Collapse
Affiliation(s)
- Carlo Galli
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Giuseppe Pedrazzi
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Stefano Guizzardi
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| |
Collapse
|
50
|
Pi Y, Liang H, Yu Q, Yin Y, Xu H, Lei Y, Han Z, Tian J. Low‑frequency pulsed electromagnetic field inhibits RANKL‑induced osteoclastic differentiation in RAW264.7 cells by scavenging reactive oxygen species. Mol Med Rep 2019; 19:4129-4136. [PMID: 30942408 PMCID: PMC6470919 DOI: 10.3892/mmr.2019.10079] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Accepted: 02/15/2019] [Indexed: 12/28/2022] Open
Abstract
Bone homeostasis is a dynamic balance maintained by bone formation and resorption. An increase in the number and activity of osteoclasts leads to excessive bone resorption, which in turn results in bone disease, including osteoporosis. Therefore, inhibiting the differentiation and activity of osteoclasts is important for maintaining bone mass. Several studies have revealed that the use of a low-frequency pulsed electromagnetic field (PEMF) is an effective method to treat osteoporosis. However, its exact mechanism remains to be fully clarified. Therefore, the present study was designed to examine the effects that PEMF exerts on receptor activator of nuclear factor-κB ligand (RANKL)-induced osteoclastogenesis and intracellular reactive oxygen species (ROS) production in RAW264.7 cells. The viability of cells was determined using a Cell Counting Kit-8 assay, and gene and protein expression were investigated via reverse transcription-quantitative polymerase chain reaction and western blot analyses. Furthermore, microscopy was performed to detect the levels of intracellular ROS and tartrate-resistant acid phosphatase (TRAP). Following the culture of RAW264.7 cells with RANKL (50 ng/ml) for 4 days (3 h/day) under PEMF (75 Hz, 1 mt) exposure, it was observed that PEMF had an inhibitory effect on RANKL-induced osteoclastic differentiation. Multinucleated osteoclast formation, the activity of TRAP and the expression of osteoclastogenesis-associated genes, including cathepsin K, nuclear factor of activated T cells cytoplasmic 1 and TRAP, were significantly reduced by PEMF. Furthermore, PEMF effectively decreased the generation of intracellular ROS during osteoclastic differentiation. In addition, the results demonstrated that ROS are the key factor in osteoclast differentiation and formation. Reducing intracellular ROS with diphenylene-iodonium chloride significantly inhibited RANKL-induced osteoclast differentiation. Taken together, the results of the present study demonstrated that PEMF may inhibit RANKL-induced osteoclastogenesis by scavenging intracellular ROS. These results may provide the groundwork for future PEMF clinical applications in osteoclast-associated bone disease.
Collapse
Affiliation(s)
- Ying Pi
- Department of Orthopedics, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510280, P.R. China
| | - Haifeng Liang
- Department of Human Anatomy, Basic Medical College, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Qiang Yu
- Department of Human Anatomy, Basic Medical College, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Yukun Yin
- Department of Human Anatomy, Basic Medical College, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Haixia Xu
- Department of Human Anatomy, Basic Medical College, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Yutian Lei
- Department of Human Anatomy, Basic Medical College, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Zhongyu Han
- Department of Human Anatomy, Basic Medical College, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Jing Tian
- Department of Orthopedics, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510280, P.R. China
| |
Collapse
|