1
|
Tota M, Jonderko L, Witek J, Novickij V, Kulbacka J. Cellular and Molecular Effects of Magnetic Fields. Int J Mol Sci 2024; 25:8973. [PMID: 39201657 PMCID: PMC11354277 DOI: 10.3390/ijms25168973] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 08/09/2024] [Accepted: 08/14/2024] [Indexed: 09/02/2024] Open
Abstract
Recently, magnetic fields (MFs) have received major attention due to their potential therapeutic applications and biological effects. This review provides a comprehensive analysis of the cellular and molecular impacts of MFs, with a focus on both in vitro and in vivo studies. We investigate the mechanisms by which MFs influence cell behavior, including modifications in gene expression, protein synthesis, and cellular signaling pathways. The interaction of MFs with cellular components such as ion channels, membranes, and the cytoskeleton is analyzed, along with their effects on cellular processes like proliferation, differentiation, and apoptosis. Molecular insights are offered into how MFs modulate oxidative stress and inflammatory responses, which are pivotal in various pathological conditions. Furthermore, we explore the therapeutic potential of MFs in regenerative medicine, cancer treatment, and neurodegenerative diseases. By synthesizing current findings, this article aims to elucidate the complex bioeffects of MFs, thereby facilitating their optimized application in medical and biotechnological fields.
Collapse
Affiliation(s)
- Maciej Tota
- Student Research Group № K148, Faculty of Medicine, Wroclaw Medical University, 50-367 Wroclaw, Poland;
| | - Laura Jonderko
- Student Research Group № K148, Faculty of Pharmacy, Wroclaw Medical University, 50-367 Wroclaw, Poland; (L.J.); (J.W.)
| | - Julia Witek
- Student Research Group № K148, Faculty of Pharmacy, Wroclaw Medical University, 50-367 Wroclaw, Poland; (L.J.); (J.W.)
| | - Vitalij Novickij
- Institute of High Magnetic Fields, Vilnius Gediminas Technical University, LT-03227 Vilnius, Lithuania;
- Department of Immunology, State Research Institute Centre for Innovative Medicine, Santariškių 5, LT-08410 Vilnius, Lithuania
| | - Julita Kulbacka
- Department of Immunology, State Research Institute Centre for Innovative Medicine, Santariškių 5, LT-08410 Vilnius, Lithuania
- Department of Molecular and Cellular Biology, Faculty of Pharmacy, Wroclaw Medical University, 50-367 Wrocław, Poland
| |
Collapse
|
2
|
Chen B, Du G, Li K, Wang Y, Shi P, Li J, Bai Y. Properties of Myofibrillar Protein in Frozen Pork Improved through pH-Shifting Treatments: The Impact of Magnetic Field. Foods 2024; 13:1988. [PMID: 38998495 PMCID: PMC11241723 DOI: 10.3390/foods13131988] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 06/07/2024] [Accepted: 06/10/2024] [Indexed: 07/14/2024] Open
Abstract
The present study demonstrates the effects of pH-shifting treatments and magnetic field-assisted pH-shifting treatments on the properties of myofibrillar protein (MP) in frozen meat. The solubility results indicate that the pH-shifting treatments increased the solubility of MP from 16.8% to a maximum of 21.0% (pH 9). The values of surface hydrophobicity and protein particle size distribution indicate that the pH-shifting treatment effectively inhibited protein aggregation through electrostatic interactions. However, under higher pH conditions (pH 10, 11), the treatments assisted by the magnetic field increased the degree of aggregation. The total thiol content and SDS-PAGE results further suggest that the magnetic field-assisted pH-shifting treatment accelerated the formation of covalent bonds among MPs under the alkaline environment. The results of the Differential Scanning Calorimetry (DSC) and protein secondary structure analysis indicate that the magnetic field promoted the unfolding of protein structures in an alkaline environment, markedly reducing the effective pH levels of pH-shifting. Electron paramagnetic resonance (EPR) data indicate that the phenomenon might be associated with the increased concentration of free radicals caused by the magnetic field treatment. In summary, the application of magnetic field-assisted pH-shifting treatments could emerge as a potent and promising strategy to improve the protein properties in frozen meat.
Collapse
Affiliation(s)
- Bo Chen
- College of Food and Bioengineering, Zhengzhou University of Light Industry, Zhengzhou 450001, China; (B.C.)
- Key Laboratory of Cold Chain Food Processing and Safety Control, Ministry of Education, Zhengzhou 450001, China
| | - Gaoang Du
- College of Food and Bioengineering, Zhengzhou University of Light Industry, Zhengzhou 450001, China; (B.C.)
- Key Laboratory of Cold Chain Food Processing and Safety Control, Ministry of Education, Zhengzhou 450001, China
| | - Ke Li
- College of Food and Bioengineering, Zhengzhou University of Light Industry, Zhengzhou 450001, China; (B.C.)
- Key Laboratory of Cold Chain Food Processing and Safety Control, Ministry of Education, Zhengzhou 450001, China
| | - Yu Wang
- College of Food and Bioengineering, Zhengzhou University of Light Industry, Zhengzhou 450001, China; (B.C.)
- Key Laboratory of Cold Chain Food Processing and Safety Control, Ministry of Education, Zhengzhou 450001, China
| | - Panpan Shi
- College of Food and Bioengineering, Zhengzhou University of Light Industry, Zhengzhou 450001, China; (B.C.)
- Key Laboratory of Cold Chain Food Processing and Safety Control, Ministry of Education, Zhengzhou 450001, China
| | - Junguang Li
- College of Food and Bioengineering, Zhengzhou University of Light Industry, Zhengzhou 450001, China; (B.C.)
- Key Laboratory of Cold Chain Food Processing and Safety Control, Ministry of Education, Zhengzhou 450001, China
| | - Yanhong Bai
- College of Food and Bioengineering, Zhengzhou University of Light Industry, Zhengzhou 450001, China; (B.C.)
- Key Laboratory of Cold Chain Food Processing and Safety Control, Ministry of Education, Zhengzhou 450001, China
| |
Collapse
|
3
|
Zafari J, Rastegar-Pouyani N, Javani Jouni F, Najjar N, Azarshin SZ, Jafarzadeh E, Abdolmaleki P, Hoseini Shirazi F. Static magnetic field reduces cisplatin resistance via increasing apoptosis pathways and genotoxicity in cancer cell lines. Sci Rep 2024; 14:5792. [PMID: 38461218 PMCID: PMC10924938 DOI: 10.1038/s41598-024-56605-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Accepted: 03/08/2024] [Indexed: 03/11/2024] Open
Abstract
Cisplatin is a chemotherapy drug widely used in cancer treatment. Alongside its clinical benefits, however, it may inflict intolerable toxicity and other adverse effects on healthy tissues. Due to the limitation of administering a high dose of cisplatin as well as cancer drug resistance, it is necessary to utilize new methods optimizing treatment modalities through both higher therapeutic efficacy and reduced administered doses of radiation and drugs. In this study, sensitive (A2780) and resistant (A2780CP) ovarian carcinoma cells underwent treatment with cisplatin + static magnetic field (SMF). First, the levels of genotoxicity after treatment were evaluated by Comet assay. Then, cell cycle analysis and apoptosis assay were conducted by a flow cytometer. Lastly, the expression levels of genes involved in apoptosis and cellular drug uptake were investigated by PCR. After treating different groups of cells for 24, 48, and 96 h, the co-treatment of SMF and cisplatin as a combination managed to increase the amount of DNA damage in both sensitive and resistant cell lines. A considerable increase in mortality of cells was also observed mostly in the form of apoptosis, which was caused by inhibition of the cell cycle. The combination also increased the expression levels of apoptotic genes, namely P53 and P21; however, it did not have much effect on the expression levels of BCL2. Besides, the levels of CTR1 gene expression increased significantly in the groups receiving the aforementioned combination. Our study suggests that the combination of cisplatin + SMF might have clinical potential which needs further investigations through future studies.
Collapse
Affiliation(s)
- Jaber Zafari
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| | - Nima Rastegar-Pouyani
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Javani Jouni
- Department of Biochemistry and Biophysics, Faculty of Advanced Sciences and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Nabaa Najjar
- Laser Application in Medical Sciences Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Seyedeh Zohreh Azarshin
- Laser Application in Medical Sciences Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Emad Jafarzadeh
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Parviz Abdolmaleki
- Department of Biophysics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Farshad Hoseini Shirazi
- Pharmaceutical Sciences Research Center and Department of Toxicology and Pharmacology, Faculty of Pharmacy, Shahid Behesthi University of Medical Sciences, Tehran, Iran
| |
Collapse
|
4
|
Xie W, Song C, Guo R, Zhang X. Static magnetic fields in regenerative medicine. APL Bioeng 2024; 8:011503. [PMID: 38486824 PMCID: PMC10939708 DOI: 10.1063/5.0191803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2023] [Accepted: 02/19/2024] [Indexed: 03/17/2024] Open
Abstract
All organisms on Earth live in the weak but ubiquitous geomagnetic field. Human beings are also exposed to magnetic fields generated by multiple sources, ranging from permanent magnets to magnetic resonance imaging (MRI) in hospitals. It has been shown that different magnetic fields can generate various effects on different tissues and cells. Among them, stem cells appear to be one of the most sensitive cell types to magnetic fields, which are the fundamental units of regenerative therapies. In this review, we focus on the bioeffects of static magnetic fields (SMFs), which are related to regenerative medicine. Most reports in the literature focus on the influence of SMF on bone regeneration, wound healing, and stem cell production. Multiple aspects of the cellular events, including gene expression, cell signaling pathways, reactive oxygen species, inflammation, and cytoskeleton, have been shown to be affected by SMFs. Although no consensus yet, current evidence indicates that moderate and high SMFs could serve as a promising physical tool to promote bone regeneration, wound healing, neural differentiation, and dental regeneration. All in vivo studies of SMFs on bone regeneration and wound healing have shown beneficial effects, which unravel the great potential of SMFs in these aspects. More mechanistic studies, magnetic field parameter optimization, and clinical investigations on human bodies will be imperative for the successful clinical applications of SMFs in regenerative medicine.
Collapse
Affiliation(s)
| | - Chao Song
- High Magnetic Field Laboratory, Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, HFIPS, Chinese Academy of Sciences, Hefei, Anhui 230031, China
| | - Ruowen Guo
- High Magnetic Field Laboratory, Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, HFIPS, Chinese Academy of Sciences, Hefei, Anhui 230031, China
| | - Xin Zhang
- Author to whom correspondence should be addressed:. Tel.: 86–551-65593356
| |
Collapse
|
5
|
Guo Z, Zhu J, Qin G, Jia Y, Liu Z, Yang N, Guo R. Static Magnetic Fields Promote Generation of Muscle Lineage Cells from Pluripotent Stem Cells and Myoblasts. Stem Cell Rev Rep 2023; 19:1402-1414. [PMID: 37000377 DOI: 10.1007/s12015-023-10535-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/21/2023] [Indexed: 04/01/2023]
Abstract
Static magnetic fields (SMFs) exhibit numerous biological effects and regulate the proliferation and differentiation of several adult stem cells. However, the role of SMFs in the self-renewal maintenance and developmental potential of pluripotent embryonic stem cells (ESCs) remains largely uninvestigated. Here, we show that SMFs promote the expression of the core pluripotent markers Sox2 and SSEA-1. Furthermore, SMFs facilitate the differentiation of ESCs into cardiomyocytes and skeletal muscle cells. Consistently, transcriptome analysis reveals that muscle lineage differentiation and skeletal system specification of ESCs are remarkably strengthened by SMF stimuli. Additionally, when treated with SMFs, C2C12 myoblasts exhibit an increased proliferation rate, improved expression of skeletal muscle markers and elevated myogenic differentiation capacity compared with control cells. Together, our data show that SMFs effectively promote muscle cell generation from pluripotent stem cells and myoblasts. The noninvasive and convenient physical stimuli can be used to increase the production of muscle cells in regenerative medicine and the manufacture of cultured meat in cellular agriculture.
Collapse
Affiliation(s)
- Zhaoyuan Guo
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China
| | - Jiahao Zhu
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China
| | - Guanyu Qin
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China
| | - Yumei Jia
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China
| | - Zheng Liu
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China
| | - Na Yang
- School of Food Science and Technology, Jiangnan University, 1800 Lihu Road, Wuxi, 214122, China
- INDUC Scientific Co., Ltd, No. 28-132 Jinshan North Photoelectric Science and Technology Park, Wuxi, 214000, China
| | - Renpeng Guo
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China.
| |
Collapse
|
6
|
Ribeiro TP, Flores M, Madureira S, Zanotto F, Monteiro FJ, Laranjeira MS. Magnetic Bone Tissue Engineering: Reviewing the Effects of Magnetic Stimulation on Bone Regeneration and Angiogenesis. Pharmaceutics 2023; 15:1045. [PMID: 37111531 PMCID: PMC10143200 DOI: 10.3390/pharmaceutics15041045] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 03/07/2023] [Accepted: 03/21/2023] [Indexed: 04/29/2023] Open
Abstract
Bone tissue engineering emerged as a solution to treat critical bone defects, aiding in tissue regeneration and implant integration. Mainly, this field is based on the development of scaffolds and coatings that stimulate cells to proliferate and differentiate in order to create a biologically active bone substitute. In terms of materials, several polymeric and ceramic scaffolds have been developed and their properties tailored with the objective to promote bone regeneration. These scaffolds usually provide physical support for cells to adhere, while giving chemical and physical stimuli for cell proliferation and differentiation. Among the different cells that compose the bone tissue, osteoblasts, osteoclasts, stem cells, and endothelial cells are the most relevant in bone remodeling and regeneration, being the most studied in terms of scaffold-cell interactions. Besides the intrinsic properties of bone substitutes, magnetic stimulation has been recently described as an aid in bone regeneration. External magnetic stimulation induced additional physical stimulation in cells, which in combination with different scaffolds, can lead to a faster regeneration. This can be achieved by external magnetic fields alone, or by their combination with magnetic materials such as nanoparticles, biocomposites, and coatings. Thus, this review is designed to summarize the studies on magnetic stimulation for bone regeneration. While providing information regarding the effects of magnetic fields on cells involved in bone tissue, this review discusses the advances made regarding the combination of magnetic fields with magnetic nanoparticles, magnetic scaffolds, and coatings and their subsequent influence on cells to reach optimal bone regeneration. In conclusion, several research works suggest that magnetic fields may play a role in regulating the growth of blood vessels, which are critical for tissue healing and regeneration. While more research is needed to fully understand the relationship between magnetism, bone cells, and angiogenesis, these findings promise to develop new therapies and treatments for various conditions, from bone fractures to osteoporosis.
Collapse
Affiliation(s)
- Tiago P. Ribeiro
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
- INEB-Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
- FEUP-Faculdade de Engenharia, Universidade do Porto, Rua Dr. Roberto Frias, s/n, 4200-465 Porto, Portugal
- Porto Comprehensive Cancer Center Raquel Seruca (P.CCC), Rua Dr. António Bernardino de Almeida, 4200-072 Porto, Portugal
| | - Miguel Flores
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
- INEB-Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
- FEUP-Faculdade de Engenharia, Universidade do Porto, Rua Dr. Roberto Frias, s/n, 4200-465 Porto, Portugal
| | - Sara Madureira
- Escola Superior de Biotecnologia, CBQF-Centro de Biotecnologia e Química Fina–Laboratório Associado, Universidade Católica Portuguesa, Rua Diogo Botelho 1327, 4169-005 Porto, Portugal
- Centro de Investigação Interdisciplinar em Saúde, Instituto de Ciências da Saúde, Universidade Católica Portuguesa, Rua Diogo Botelho 1327, 4169-005 Porto, Portugal
| | - Francesca Zanotto
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
- INEB-Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
- Department of Information Engineering, University of Padua, Via Gradenigo 6/b, 35131 Padova, Italy
| | - Fernando J. Monteiro
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
- INEB-Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
- FEUP-Faculdade de Engenharia, Universidade do Porto, Rua Dr. Roberto Frias, s/n, 4200-465 Porto, Portugal
- Porto Comprehensive Cancer Center Raquel Seruca (P.CCC), Rua Dr. António Bernardino de Almeida, 4200-072 Porto, Portugal
| | - Marta S. Laranjeira
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
- INEB-Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
- Porto Comprehensive Cancer Center Raquel Seruca (P.CCC), Rua Dr. António Bernardino de Almeida, 4200-072 Porto, Portugal
| |
Collapse
|
7
|
Phenolic acids and a static magnetic field change the expression of transforming growth factor β isoforms in amelanotic melanoma cells. Mol Biol Rep 2023; 50:4207-4216. [PMID: 36899279 PMCID: PMC10147755 DOI: 10.1007/s11033-023-08336-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2022] [Accepted: 02/14/2023] [Indexed: 03/12/2023]
Abstract
BACKGROUND Melanoma is an aggressive type of cancer that can metastasize to numerous other organs. TGFβ is one of the key signaling pathways in melanoma progression. Previous studies on various types of cancer have shown that both: polyphenols and a static magnetic field (SMF) can be potential chemopreventive/therapeutic agents. Therefore, the aim of the study was to evaluate the effect of a SMF and selected polyphenols on the transcriptional activity of TGFβ genes in melanoma cells. METHODS AND RESULTS Experiments were performed on the C32 cell line treated with caffeic or chlorogenic acids, and with simultaneous exposure to a moderate-strength SMF. The RT-qPCR method was used to determine the mRNA level of genes encoding the TGFβ isoforms and their receptors. The concentration of the TGFβ1 and TGFβ2 proteins were also measured in the cell culture supernates. The first response of C32 melanoma cells to both factors is the reduction of TGFβ levels. Then, mRNA level of these molecules returned to values close to pre-treatment level by the end of experiment. CONCLUSION Our study results demonstrate the potential of polyphenols and a moderate-strength SMF to support cancer therapy by altering TGFβ expression, which is a very promising topic for the diagnosis and treatment of melanoma.
Collapse
|
8
|
Wang J, Shang P. Static magnetic field: A potential tool of controlling stem cells fates for stem cell therapy in osteoporosis. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2023; 178:91-102. [PMID: 36596343 DOI: 10.1016/j.pbiomolbio.2022.12.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/01/2022] [Revised: 12/10/2022] [Accepted: 12/29/2022] [Indexed: 01/01/2023]
Abstract
Osteoporosis is a kind of bone diseases characterized by dynamic imbalance of bone formation and bone absorption, which is prone to fracture, and seriously endangers human health. At present, there is a lack of highly effective drugs for it, and the existing measures all have some side effects. In recent years, mesenchymal stem cell therapy has brought a certain hope for osteoporosis, while shortcomings such as homing difficulty and unstable therapeutic effects limit its application widely. Therefore, it is extremely urgent to find effective and reliable means/drugs for adjuvant stem cell therapy or develop new research techniques. It has been reported that static magnetic fields(SMFs) has a certain alleviating and therapeutic effect on varieties of bone diseases, also promotes the proliferation and osteogenic differentiation of mesenchymal stem cells derived from different tissues to a certain extent. Basing on the above background, this article focuses on the key words "static/constant magnetic field, mesenchymal stem cell, osteoporosis", combined literature and relevant contents were studied to look forward that SMFs has unique advantages in the treatment of osteoporosis with mesenchymal stem cells, which can be used as an application tool to promote the progress of stem cell therapy in clinical application.
Collapse
Affiliation(s)
- Jianping Wang
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi, 710072, China; Key Laboratory for Space Bioscience and Biotechnology, Institute of Special Environmental Biophysics, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi, 710072, China
| | - Peng Shang
- Research & Development Institute of Northwestern Polytechnical University in Shenzhen, Shenzhen, 518057, China; School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi, 710072, China; Key Laboratory for Space Bioscience and Biotechnology, Institute of Special Environmental Biophysics, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi, 710072, China.
| |
Collapse
|
9
|
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: 17.0] [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
|
10
|
Toda T, Ito M, Takeda JI, Masuda A, Mino H, Hattori N, Mohri K, Ohno K. Extremely low-frequency pulses of faint magnetic field induce mitophagy to rejuvenate mitochondria. Commun Biol 2022; 5:453. [PMID: 35552531 PMCID: PMC9098439 DOI: 10.1038/s42003-022-03389-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Accepted: 04/21/2022] [Indexed: 11/25/2022] Open
Abstract
Humans are frequently exposed to time-varying and static weak magnetic fields (WMF). However, the effects of faint magnetic fields, weaker than the geomagnetic field, have been scarcely reported. Here we show that extremely low-frequency (ELF)-WMF, comprised of serial pulses of 10 µT intensity at 1–8 Hz, which is three or more times weaker than the geomagnetic field, reduces mitochondrial mass to 70% and the mitochondrial electron transport chain (ETC) complex II activity to 88%. Chemical inhibition of electron flux through the mitochondrial ETC complex II nullifies the effect of ELF-WMF. Suppression of ETC complex II subsequently induces mitophagy by translocating parkin and PINK1 to the mitochondria and by recruiting LC3-II. Thereafter, mitophagy induces PGC-1α-mediated mitochondrial biogenesis to rejuvenate mitochondria. The lack of PINK1 negates the effect of ELF-WMF. Thus, ELF-WMF may be applicable for the treatment of human diseases that exhibit compromised mitochondrial homeostasis, such as Parkinson’s disease. The effect of extremely low-frequency pulses of faint magnetic field on mitochondria is investigated, where it led to reduced mitochondrial mass, membrane potential and electron transport chain activity, and induced mitophagy.
Collapse
Affiliation(s)
- Takuro Toda
- Division of Neurogenetics, Center for Neurological Diseases and Cancer, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Mikako Ito
- Division of Neurogenetics, Center for Neurological Diseases and Cancer, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Jun-Ichi Takeda
- Division of Neurogenetics, Center for Neurological Diseases and Cancer, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Akio Masuda
- Division of Neurogenetics, Center for Neurological Diseases and Cancer, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Hiroyuki Mino
- Division of Material Science, Nagoya University Graduate School of Science, Nagoya, Japan
| | | | - Kaneo Mohri
- Nagoya Industrial Science Research Institute, Nagoya, Japan
| | - Kinji Ohno
- Division of Neurogenetics, Center for Neurological Diseases and Cancer, Nagoya University Graduate School of Medicine, Nagoya, Japan.
| |
Collapse
|
11
|
Song C, Yu B, Wang J, Zhu Y, Zhang X. Effects of Moderate to High Static Magnetic Fields on Reproduction. Bioelectromagnetics 2022; 43:278-291. [PMID: 35485707 DOI: 10.1002/bem.22404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2021] [Revised: 03/09/2022] [Accepted: 04/09/2022] [Indexed: 11/08/2022]
Abstract
With the wide application of magnetic resonance imaging in hospitals and permanent magnets in household items, people have increased exposure to various types of static magnetic fields (SMFs) with moderate and high intensities, which has caused a considerable amount of public concern. Studies have shown that some aspects of gametogenesis and early embryonic development can be significantly affected by SMFs, while others have shown no effects. This review summarizes the experimental results of moderate to high-intensity SMFs (1 mT-16.7 T) on the reproductive development of different model animals, and we find that the effects of SMFs are variable depending on experimental conditions. In general, the effects of inhomogeneous SMFs seem to be more significant compared to that of homogeneous SMFs, which is likely due to magnetic forces generated by the magnetic field gradient. Moreover, some electromagnetic fields may have induced bioeffects because of nonnegligible gradient and heat effect, which are much reduced in superconducting magnets. We hope this review can provide a starting point for more in-depth analysis of various SMFs on reproduction, which is indispensable for evaluating the safety and potential applications of SMFs on living organisms in the future. © 2022 Bioelectromagnetics Society.
Collapse
Affiliation(s)
- Chao Song
- High Magnetic Field Laboratory, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, China.,University of Science and Technology of China, Hefei, China
| | - Biao Yu
- High Magnetic Field Laboratory, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, China.,University of Science and Technology of China, Hefei, China
| | - Junjun Wang
- High Magnetic Field Laboratory, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, China
| | - Yiming Zhu
- High Magnetic Field Laboratory, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, China.,Institutes of Physical Science and Information Technology, Anhui University, Hefei, China
| | - Xin Zhang
- High Magnetic Field Laboratory, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, China.,University of Science and Technology of China, Hefei, China.,Institutes of Physical Science and Information Technology, Anhui University, Hefei, China.,International Magnetobiology Frontier Research Center (iMFRC), Science Island, Hefei, China
| |
Collapse
|
12
|
Tian L, Luo Y, Zhan A, Ren J, Qin H, Pan Y. Hypomagnetic Field Induces the Production of Reactive Oxygen Species and Cognitive Deficits in Mice Hippocampus. Int J Mol Sci 2022; 23:ijms23073622. [PMID: 35408982 PMCID: PMC8998670 DOI: 10.3390/ijms23073622] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 03/20/2022] [Accepted: 03/22/2022] [Indexed: 12/14/2022] Open
Abstract
Previous studies have found that hypomagnetic field (HMF) exposure impairs cognition behaviors in animals; however, the underlying neural mechanisms of cognitive dysfunction are unclear. The hippocampus plays important roles in magnetoreception, memory, and spatial navigation in mammals. Therefore, the hippocampus may be the key region in the brain to reveal its neural mechanisms. We recently reported that long-term HMF exposure impairs adult hippocampal neurogenesis and cognition through reducing endogenous reactive oxygen species (ROS) levels in adult neural stem cells that are confined in the subgranular zone (SGZ) of the hippocampus. In addition to adult neural stem cells, the redox state of other cells in the hippocampus is also an important factor affecting the functions of the hippocampus. However, it is unclear whether and how long-term HMF exposure affects ROS levels in the entire hippocampus (i.e., the dentate gyrus (DG) and ammonia horn (CA) regions). Here, we demonstrate that male C57BL/6J mice exposed to 8-week HMF exhibit cognitive impairments. We then found that the ROS levels of the hippocampus were significantly higher in these HMF-exposed mice than in the geomagnetic field (GMF) group. PCR array analysis revealed that the elevated ROS levels were due to HMF-regulating genes that maintain the redox balance in vivo, such as Nox4, Gpx3. Since high levels of ROS may cause hippocampal oxidative stress, we suggest that this is another reason why HMF exposure induces cognitive impairment, besides the hippocampal neurogenesis impairments. Our study further demonstrates that GMF plays an important role in maintaining hippocampal function by regulating the appropriate endogenous ROS levels.
Collapse
Affiliation(s)
- Lanxiang Tian
- Biogeomagnetism Group, Key Laboratory of Earth and Planetary Physics, Institute of Geology and Geophysics, Chinese Academy of Sciences, Beijing 100029, China; (Y.L.); (A.Z.); (J.R.); (Y.P.)
- Innovation Academy for Earth Science, Chinese Academy of Sciences, Beijing 100029, China;
- The Paleomagnetism and Geochronology Laboratory, Institute of Geology and Geophysics, Chinese Academy of Sciences, Beijing 100029, China
- Correspondence:
| | - Yukai Luo
- Biogeomagnetism Group, Key Laboratory of Earth and Planetary Physics, Institute of Geology and Geophysics, Chinese Academy of Sciences, Beijing 100029, China; (Y.L.); (A.Z.); (J.R.); (Y.P.)
- Innovation Academy for Earth Science, Chinese Academy of Sciences, Beijing 100029, China;
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Aisheng Zhan
- Biogeomagnetism Group, Key Laboratory of Earth and Planetary Physics, Institute of Geology and Geophysics, Chinese Academy of Sciences, Beijing 100029, China; (Y.L.); (A.Z.); (J.R.); (Y.P.)
- Innovation Academy for Earth Science, Chinese Academy of Sciences, Beijing 100029, China;
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jie Ren
- Biogeomagnetism Group, Key Laboratory of Earth and Planetary Physics, Institute of Geology and Geophysics, Chinese Academy of Sciences, Beijing 100029, China; (Y.L.); (A.Z.); (J.R.); (Y.P.)
- Innovation Academy for Earth Science, Chinese Academy of Sciences, Beijing 100029, China;
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Huafeng Qin
- Innovation Academy for Earth Science, Chinese Academy of Sciences, Beijing 100029, China;
- The Paleomagnetism and Geochronology Laboratory, Institute of Geology and Geophysics, Chinese Academy of Sciences, Beijing 100029, China
| | - Yongxin Pan
- Biogeomagnetism Group, Key Laboratory of Earth and Planetary Physics, Institute of Geology and Geophysics, Chinese Academy of Sciences, Beijing 100029, China; (Y.L.); (A.Z.); (J.R.); (Y.P.)
- Innovation Academy for Earth Science, Chinese Academy of Sciences, Beijing 100029, China;
- The Paleomagnetism and Geochronology Laboratory, Institute of Geology and Geophysics, Chinese Academy of Sciences, Beijing 100029, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
13
|
An Open Question: Is Non-Ionizing Radiation a Tool for Controlling Apoptosis-Induced Proliferation? Int J Mol Sci 2021; 22:ijms222011159. [PMID: 34681819 PMCID: PMC8537877 DOI: 10.3390/ijms222011159] [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: 09/24/2021] [Accepted: 10/08/2021] [Indexed: 12/24/2022] Open
Abstract
Non-ionizing radiation is commonly used in the clinical setting, despite its known ability to trigger oxidative stress and apoptosis, which can lead to damage and cell death. Although induction of cell death is typically considered harmful, apoptosis can also be beneficial in the right context. For example, cell death can serve as the signal for new tissue growth, such as in apoptosis-induced proliferation. Recent data has shown that exposure to non-ionizing radiation (such as weak static magnetic fields, weak radiofrequency magnetic fields, and weak electromagnetic fields) is able to modulate proliferation, both in cell culture and in living organisms (for example during tissue regeneration). This occurs via in vivo changes in the levels of reactive oxygen species (ROS), which are canonical activators of apoptosis. This review will describe the literature that highlights the tantalizing possibility that non-ionizing radiation could be used to manipulate apoptosis-induced proliferation to either promote growth (for regenerative medicine) or inhibit it (for cancer therapies). However, as uncontrolled growth can lead to tumorigenesis, much more research into this exciting and developing area is needed in order to realize its promise.
Collapse
|
14
|
Sharma AK, Sah S, Singla SK, Chauhan MS, Manik RS, Palta P. Exposure to Pulsed Electromagnetic Fields Improves the Developmental Competence and Quality of Somatic Cell Nuclear Transfer Buffalo ( Bubalus bubalis) Embryos Produced Using Fibroblast Cells and Alters Their Epigenetic Status and Gene Expression. Cell Reprogram 2021; 23:304-315. [PMID: 34597162 DOI: 10.1089/cell.2021.0028] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
We examined the effects of treatment with pulsed electromagnetic fields (PEMFs) on cumulus cells and buffalo somatic cell nuclear transfer (SCNT) embryos. PEMF treatment (30 μT for 3 hours) of cumulus cells increased (p < 0.05) the relative cell viability and cell proliferation and the expression level of OCT4, NANOG, SOX2, P53, CCNB1, and GPX, but decreased (p < 0.05) that of DNMT1, DNMT3a, GSK3b, and BAX, whereas the expression level of DNMT3b, GLUT1, BCL2, CASPASE3, SOD1, and CATALASE was not affected. PEMF treatment of SCNT embryos at the beginning of in vitro culture increased (p < 0.05) the blastocyst rate (51.4% ± 1.36% vs. 42.8% ± 1.29%) and decreased (p < 0.01) the apoptotic index to the level in in vitro fertilization blastocysts, but did not significantly alter the total cell number and the inner cell mass:trophectoderm cell number ratio of blastocysts compared to the controls. PEMF treatment increased the expression level of NANOG, SOX2, CDX2, GLUT1, P53, and BCL2 and decreased that of BAX, CASPASE3, GSK3b, and HSP70, but not OCT4, DNMT1, DNMT3a, DNMT3b, HDAC1, and CCNB1 in blastocysts. It increased (p < 0.001) the global level of H3K27me3 but not H3K18ac. These results suggest that PEMF treatment of SCNT embryos improves their developmental competence, reduces the level of apoptosis, and alters the expression level of several important genes related to pluripotency, apoptosis, metabolism, and stress.
Collapse
Affiliation(s)
- Aditya Kumar Sharma
- Animal Biotechnology Centre, ICAR-National Dairy Research Institute, Karnal, India
| | - Shrutika Sah
- Animal Biotechnology Centre, ICAR-National Dairy Research Institute, Karnal, India
| | - Suresh Kumar Singla
- Animal Biotechnology Centre, ICAR-National Dairy Research Institute, Karnal, India
| | | | - Radhey Shyam Manik
- Animal Biotechnology Centre, ICAR-National Dairy Research Institute, Karnal, India
| | - Prabhat Palta
- Animal Biotechnology Centre, ICAR-National Dairy Research Institute, Karnal, India.,Embryo Biotechnology Laboratory, Animal Biotechnology Centre, ICAR-National Dairy Research Institute, Karnal, India
| |
Collapse
|
15
|
Zhao B, Yu T, Wang S, Che J, Zhou L, Shang P. Static Magnetic Field (0.2-0.4 T) Stimulates the Self-Renewal Ability of Osteosarcoma Stem Cells Through Autophagic Degradation of Ferritin. Bioelectromagnetics 2021; 42:371-383. [PMID: 34082485 DOI: 10.1002/bem.22352] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 04/07/2021] [Accepted: 05/17/2021] [Indexed: 12/25/2022]
Abstract
Static magnetic field (SMF) can alter cell fate decisions in many ways. However, the effects of SMF on cancer stem cells (CSCs) are little-known. In this particular study, we evaluate the biological effect of moderate-intensity SMF on osteosarcoma stem cells (OSCs) and try to clarify the underlying mechanisms of action. First, we demonstrated that prolonged exposure to SMF induced the proliferation and tumorsphere formation in K7M2 and MG63 OSCs. Moreover, SMF promoted the release of ferrous iron (Fe2+ ) and provoked reactive oxygen species (ROS) in OSCs. Interestingly, SMF evidently triggered the autophagic degradation of ferritin, which is characterized by the activation of microtubule-associated protein 1 light chain 3 (LC3) and nuclear receptor co-activator 4 (NCOA4), and downregulation of ferritin heavy chain 1 (FTH1) in OSCs. Particularly, the colony-forming ability of K7M2 OSCs promoted by SMF was obviously abolished by using a small interfering RNA (siRNA) against NCOA4. Finally, treatment of the tumor-bearing mice with SMF did not affect the tumor volume or tumor mass, nor pulmonary metastasis of K7M2 OSCs, but the SMF-treated K7M2 OSCs caused a preference of pulmonary metastasis in a mouse model, which suggested that SMF might induce the metastatic characteristic of OSCs. Consequently, this paper demonstrates for the first time that the cumulative SMF exposure promoted the self-renewal ability of OSCs via autophagic degradation of ferritin, implying that ferritinophagy may be a potential molecular target for cancer. © 2021 Bioelectromagnetics Society.
Collapse
Affiliation(s)
- Bin Zhao
- Research & Development Institute of Northwestern Polytechnical University in Shenzhen, Shenzhen, China.,School of Life Science, Northwestern Polytechnical University, Xi'an, China.,Key Laboratory for Space Bioscience and Biotechnology, Institute of Special Environmental Biophysics, Northwestern Polytechnical University, Xi'an, China
| | - Tongyao Yu
- Research & Development Institute of Northwestern Polytechnical University in Shenzhen, Shenzhen, China.,School of Life Science, Northwestern Polytechnical University, Xi'an, China.,Key Laboratory for Space Bioscience and Biotechnology, Institute of Special Environmental Biophysics, Northwestern Polytechnical University, Xi'an, China
| | - Shenghang Wang
- Research & Development Institute of Northwestern Polytechnical University in Shenzhen, Shenzhen, China.,School of Life Science, Northwestern Polytechnical University, Xi'an, China.,Key Laboratory for Space Bioscience and Biotechnology, Institute of Special Environmental Biophysics, Northwestern Polytechnical University, Xi'an, China
| | - Jingmin Che
- Research & Development Institute of Northwestern Polytechnical University in Shenzhen, Shenzhen, China.,School of Life Science, Northwestern Polytechnical University, Xi'an, China.,Key Laboratory for Space Bioscience and Biotechnology, Institute of Special Environmental Biophysics, Northwestern Polytechnical University, Xi'an, China
| | - Liangfu Zhou
- Research & Development Institute of Northwestern Polytechnical University in Shenzhen, Shenzhen, China.,School of Life Science, Northwestern Polytechnical University, Xi'an, China.,Key Laboratory for Space Bioscience and Biotechnology, Institute of Special Environmental Biophysics, Northwestern Polytechnical University, Xi'an, China
| | - Peng Shang
- Research & Development Institute of Northwestern Polytechnical University in Shenzhen, Shenzhen, China.,Key Laboratory for Space Bioscience and Biotechnology, Institute of Special Environmental Biophysics, Northwestern Polytechnical University, Xi'an, China
| |
Collapse
|
16
|
Jedrzejczak-Silicka M, Kordas M, Konopacki M, Rakoczy R. Modulation of Cellular Response to Different Parameters of the Rotating Magnetic Field (RMF)-An In Vitro Wound Healing Study. Int J Mol Sci 2021; 22:5785. [PMID: 34071384 PMCID: PMC8199476 DOI: 10.3390/ijms22115785] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 05/24/2021] [Accepted: 05/24/2021] [Indexed: 12/23/2022] Open
Abstract
Since the effect of MFs (magnetic fields) on various biological systems has been studied, different results have been obtained from an insignificant effect of weak MFs on the disruption of the circadian clock system. On the other hand, magnetic fields, electromagnetic fields, or electric fields are used in medicine. The presented study was conducted to determine whether a low-frequency RMF (rotating magnetic field) with different field parameters could evoke the cellular response in vitro and is possible to modulate the cellular response. The cellular metabolic activity, ROS and Ca2+ concentration levels, wound healing assay, and gene expression analyses were conducted to evaluate the effect of RMF. It was shown that different values of magnetic induction (B) and frequency (f) of RMF evoke a different response of cells, e.g., increase in the general metabolic activity may be associated with the increasing of ROS levels. The lower intracellular Ca2+ concentration (for 50 Hz) evoked the inability of cells to wound closure. It can be stated that the subtle balance in the ROS level is crucial in the wound for the effective healing process, and it is possible to modulate the cellular response to the RMF in the context of an in vitro wound healing.
Collapse
Affiliation(s)
- Magdalena Jedrzejczak-Silicka
- Laboratory of Cytogenetics, West Pomeranian University of Technology in Szczecin, Klemensa Janickiego 29, 71-270 Szczecin, Poland;
| | - Marian Kordas
- Faculty of Chemical Technology and Engineering, West Pomeranian University of Technology in Szczecin, Piastow Avenue 42, 71-065 Szczecin, Poland; (M.K.); (M.K.)
| | - Maciej Konopacki
- Faculty of Chemical Technology and Engineering, West Pomeranian University of Technology in Szczecin, Piastow Avenue 42, 71-065 Szczecin, Poland; (M.K.); (M.K.)
| | - Rafał Rakoczy
- Faculty of Chemical Technology and Engineering, West Pomeranian University of Technology in Szczecin, Piastow Avenue 42, 71-065 Szczecin, Poland; (M.K.); (M.K.)
| |
Collapse
|
17
|
Gurhan H, Bruzon R, Kandala S, Greenebaum B, Barnes F. Effects Induced by a Weak Static Magnetic Field of Different Intensities on HT-1080 Fibrosarcoma Cells. Bioelectromagnetics 2021; 42:212-223. [PMID: 33735454 DOI: 10.1002/bem.22332] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 02/20/2021] [Accepted: 03/03/2021] [Indexed: 01/07/2023]
Abstract
In this study, we investigated the effects of weak static magnetic fields (SMFs) on HT-1080 human fibrosarcoma cells. Exposures to SMFs for four consecutive days were varied from 0.5 to 600 µT for treated units, while exposures to control units were held at 45 µT. Growth rates were measured by comparing cell counts, whereas membrane potentials, mitochondrial calcium, mitochondrial superoxide (O2 - ), nitric oxide (NO), hydrogen peroxide (H2 O2 ), intercellular pH, and oxidative stress were measured by using fluorescent dyes. The relative cell growth rates vary with the angle of the SMFs. Increases in the magnitude of the SMFs increased concentrations of mitochondrial calcium and membrane potential and decreased intracellular pH. H2 O2 , an important reactive oxygen species (ROS), increases at 100 and 200 µT, decreases at 300 and 400 µT and increases again at 500 and 600 µT. Overall, oxidative stress increases slightly with increasing SMFs, while superoxide and NO concentrations decrease. These results indicate that weak SMFs can accelerate and inhibit cell growth rates and induce alterations in ROS. Changes in ROS and oxidative stress are important for various cell functions. Calcium influx into mitochondria was one of the initial steps into the corresponding changes. Bioelectromagnetics. 2021. © 2021 Bioelectromagnetics Society.
Collapse
Affiliation(s)
- Hakki Gurhan
- Department of Electrical, Computer and Energy, University of Colorado Boulder, Boulder, Colorado
| | - Rodolfo Bruzon
- Department of Electrical, Computer and Energy, University of Colorado Boulder, Boulder, Colorado
| | - Sahithi Kandala
- Department of Electrical, Computer and Energy, University of Colorado Boulder, Boulder, Colorado
| | - Ben Greenebaum
- Department of Physics, University of Wisconsin-Parkside, Kenosha, Wisconsin
| | - Frank Barnes
- Department of Electrical, Computer and Energy, University of Colorado Boulder, Boulder, Colorado
| |
Collapse
|
18
|
Dateki M, Imamura O, Arai M, Shimizu H, Takishima K. A novel strategy of selective gene delivery by using a uniform magnetic field. Biotechnol J 2021; 16:e2000233. [PMID: 33226197 DOI: 10.1002/biot.202000233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 11/11/2020] [Indexed: 11/08/2022]
Abstract
The application of a magnetic field to enhance the transfection efficiency has been reported to be mainly dependent on the magnetic force generated by a magnetic field gradient to attract paramagnetic bead-conjugated carrier and polynucleotide complexes. This strategy has the advantage of targeting a point or an area on the culture vessel. However, it is difficult to target deeply placed tissues in vivo. Uniform magnetic field-correlated effect is applicable to such a purpose. Here, we attempted to establish a novel procedure for uniform magnetic field-dependent enhancement of transfection efficiency. We examined the effect of a 1.5 mT uniform magnetic field on cellular reactive oxygen species (ROS) level and transfection efficiency mediated by a ROS-sensitive transfection carrier. Our experimental results revealed that a 1.5 mT uniform magnetic field transiently decreased cellular ROS levels and strongly enhanced transfection efficiency mediated by polyethylenimine (PEI). The uniform magnetic field-dependent enhancement of PEI-mediated in vivo transfection was confirmed in the livers of mice. Local intensification of a uniform magnetic field in a culture dish resulted in selective gene delivery into cells on the target area. Although further examination and improvement are necessary for this procedure, our findings provide a novel option for spatial control of gene delivery.
Collapse
Affiliation(s)
- Minori Dateki
- Department of Biochemistry, National Defense Medical College, Tokorozawa, Japan
| | - Osamu Imamura
- Department of Biochemistry, National Defense Medical College, Tokorozawa, Japan
| | - Masaaki Arai
- Department of Biochemistry, National Defense Medical College, Tokorozawa, Japan
| | - Hidehisa Shimizu
- Institute of Agricultural and Life Sciences, Academic Assembly, Shimane University, Matsue, Japan
| | - Kunio Takishima
- Department of Biochemistry, National Defense Medical College, Tokorozawa, Japan
| |
Collapse
|
19
|
Zastko L, Makinistian L, Moravčíková A, Jakuš J, Belyaev I. Effect of Intermittent ELF MF on Umbilical Cord Blood Lymphocytes. Bioelectromagnetics 2020; 41:649-655. [PMID: 33190314 DOI: 10.1002/bem.22302] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 08/11/2020] [Accepted: 10/10/2020] [Indexed: 11/06/2022]
Affiliation(s)
- Lucián Zastko
- Department of Radiobiology, Cancer Research Institute, Biomedical Research Center, University Science Park for Biomedicine, Slovak Academy of Sciences, Bratislava, Slovakia
- Department of Medical Biophysics, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Martin, Slovakia
| | - Leonardo Makinistian
- Department of Physics, Instituto de Física Aplicada (INFAP), Universidad Nacional de San Luis-CONICET, San Luis, Argentina
| | - Andrea Moravčíková
- Department of Radiobiology, Cancer Research Institute, Biomedical Research Center, University Science Park for Biomedicine, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Ján Jakuš
- Department of Medical Biophysics, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Martin, Slovakia
| | - Igor Belyaev
- Department of Radiobiology, Cancer Research Institute, Biomedical Research Center, University Science Park for Biomedicine, Slovak Academy of Sciences, Bratislava, Slovakia
| |
Collapse
|
20
|
Roles of Reactive Oxygen Species in Cardiac Differentiation, Reprogramming, and Regenerative Therapies. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:2102841. [PMID: 32908625 PMCID: PMC7475763 DOI: 10.1155/2020/2102841] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Accepted: 07/22/2020] [Indexed: 12/11/2022]
Abstract
Reactive oxygen species (ROS) have been implicated in mechanisms of heart development and regenerative therapies such as the use of pluripotent stem cells. The roles of ROS mediating cell fate are dependent on the intensity of stimuli, cellular context, and metabolic status. ROS mainly act through several targets (such as kinases and transcription factors) and have diverse roles in different stages of cardiac differentiation, proliferation, and maturation. Therefore, further detailed investigation and characterization of redox signaling will help the understanding of the molecular mechanisms of ROS during different cellular processes and enable the design of targeted strategies to foster cardiac regeneration and functional recovery. In this review, we focus on the roles of ROS in cardiac differentiation as well as transdifferentiation (direct reprogramming). The potential mechanisms are discussed in regard to ROS generation pathways and regulation of downstream targets. Further methodological optimization is required for translational research in order to robustly enhance the generation efficiency of cardiac myocytes through metabolic modulations. Additionally, we highlight the deleterious effect of the host's ROS on graft (donor) cells in a paracrine manner during stem cell-based implantation. This knowledge is important for the development of antioxidant strategies to enhance cell survival and engraftment of tissue engineering-based technologies. Thus, proper timing and level of ROS generation after a myocardial injury need to be tailored to ensure the maximal efficacy of regenerative therapies and avoid undesired damage.
Collapse
|
21
|
Rodriguez ML, Werner TR, Becker B, Eschenhagen T, Hirt MN. A magnetics-based approach for fine-tuning afterload in engineered heart tissues. ACS Biomater Sci Eng 2019; 5:3663-3675. [PMID: 31637285 DOI: 10.1021/acsbiomaterials.8b01568] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Afterload plays important roles during heart development and disease progression, however, studying these effects in a laboratory setting is challenging. Current techniques lack the ability to precisely and reversibly alter afterload over time. Here, we describe a magnetics-based approach for achieving this control and present results from experiments in which this device was employed to sequentially increase afterload applied to rat engineered heart tissues (rEHTs) over a 7-day period. The contractile properties of rEHTs grown on control posts marginally increased over the observation period. The average post deflection, fractional shortening, and twitch velocities measured for afterload-affected tissues initially followed this same trend, but fell below control tissue values at high magnitudes of afterload. However, the average force, force production rate, and force relaxation rate for these rEHTs were consistently up to 3-fold higher than in control tissues. Transcript levels of hypertrophic or fibrotic markers and cell size remained unaffected by afterload, suggesting that the increased force output was not accompanied by pathological remodeling. Accordingly, the increased force output was fully reversed to control levels during a stepwise decrease in afterload over 4 hours. Afterload application did not affect systolic or diastolic tissue lengths, indicating that the afterload system was likely not a source of changes in preload strain. In summary, the afterload system developed herein is capable of fine-tuning EHT afterload while simultaneously allowing optical force measurements. Using this system, we found that small daily alterations in afterload can enhance the contractile properties of rEHTs, while larger increases can have temporary undesirable effects. Overall, these findings demonstrate the significant role that afterload plays in cardiac force regulation. Future studies with this system may allow for novel insights into the mechanisms that underlie afterload-induced adaptations in cardiac force development.
Collapse
Affiliation(s)
- Marita L Rodriguez
- Department of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246 Hamburg, Germany.,DZHK (German Center for Cardiovascular Research), Partner site Hamburg/Kiel/Lübeck, 20246 Hamburg, Germany
| | - Tessa R Werner
- Department of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246 Hamburg, Germany.,DZHK (German Center for Cardiovascular Research), Partner site Hamburg/Kiel/Lübeck, 20246 Hamburg, Germany
| | - Benjamin Becker
- Department of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246 Hamburg, Germany.,DZHK (German Center for Cardiovascular Research), Partner site Hamburg/Kiel/Lübeck, 20246 Hamburg, Germany
| | - Thomas Eschenhagen
- Department of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246 Hamburg, Germany.,DZHK (German Center for Cardiovascular Research), Partner site Hamburg/Kiel/Lübeck, 20246 Hamburg, Germany
| | - Marc N Hirt
- Department of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246 Hamburg, Germany.,DZHK (German Center for Cardiovascular Research), Partner site Hamburg/Kiel/Lübeck, 20246 Hamburg, Germany
| |
Collapse
|
22
|
Simkó M, Mattsson MO. Activation of the intracellular temperature and ROS sensor membrane protein STIM1 as a mechanism underpinning biological effects of low-level low frequency magnetic fields. Med Hypotheses 2018; 122:68-72. [PMID: 30593427 DOI: 10.1016/j.mehy.2018.10.013] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Revised: 10/16/2018] [Accepted: 10/20/2018] [Indexed: 01/22/2023]
Abstract
The biological effects of low frequency magnetic fields (LF MF) at high flux densities are well known and the underlying mechanism is established. In contrast, health associated effects at lower flux densities, which can be found in the human environment, are controversial, and no accepted interaction mechanism has been presented. Here we present a hypothesis regarding the biological aspect of interaction between LF MF and cells. We suggest that the endoplasmic reticulum (ER) membrane protein STIM1, which functions as a sensor for several cellular conditions (low Ca2+ levels, temperature increase, increased levels of oxygen radicals, hypoxia), is a candidate LF MF sensor. Such a sensor function can be either direct (via local temperature increase caused by intracellularly induced electric fields), or indirect due to responses to increased reactive oxygen species (ROS) levels. Activated STIM1 leads to downstream effects by activation of signal transduction processes and changes in gene expression leading to secondary events. The nature of these changes would be dependent on both cell type and the particular physiological state the cell displays at the time of STIM1 activation. Results from testing of this hypothesis, as suggested in this paper, would greatly assist in understanding of the possible health-related effects of low-level LF MF. This would benefit both safety assessments regarding MF exposure as well as possible use of MF in medicine. A better understanding of the biological mechanisms underpinning MF exposure effects of living matter allows the targeted use of the fields in medical applications. There are several examples already in use based on empiric and not on mechanistic knowledge. Knowledge generated from our hypothesis testing makes it possible for MF based medical applications to be optimized.
Collapse
|
23
|
Marrella A, Iafisco M, Adamiano A, Rossi S, Aiello M, Barandalla-Sobrados M, Carullo P, Miragoli M, Tampieri A, Scaglione S, Catalucci D. A combined low-frequency electromagnetic and fluidic stimulation for a controlled drug release from superparamagnetic calcium phosphate nanoparticles: potential application for cardiovascular diseases. J R Soc Interface 2018; 15:20180236. [PMID: 29997259 PMCID: PMC6073647 DOI: 10.1098/rsif.2018.0236] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Accepted: 06/19/2018] [Indexed: 01/09/2023] Open
Abstract
Alternative drug delivery approaches to treat cardiovascular diseases are currently under intense investigation. In this domain, the possibility to target the heart and tailor the amount of drug dose by using a combination of magnetic nanoparticles (NPs) and electromagnetic devices is a fascinating approach. Here, an electromagnetic device based on Helmholtz coils was generated for the application of low-frequency magnetic stimulations to manage drug release from biocompatible superparamagnetic Fe-hydroxyapatite NPs (FeHAs). Integrated with a fluidic circuit mimicking the flow of the cardiovascular environment, the device was efficient to trigger the release of a model drug (ibuprofen) from FeHAs as a function of the applied frequencies. Furthermore, the biological effects on the cardiac system of the identified electromagnetic exposure were assessed in vitro and in vivo by acute stimulation of isolated adult cardiomyocytes and in an animal model. The cardio-compatibility of FeHAs was also assessed in vitro and in an animal model. No alterations of cardiac electrophysiological properties were observed in both cases, providing the evidence that the combination of low-frequency magnetic stimulations and FeHAs might represent a promising strategy for controlled drug delivery to the failing heart.
Collapse
Affiliation(s)
- Alessandra Marrella
- National Research Council (CNR), Institute of Electronic, Computer and Telecommunications (IEIIT), via de Marini 6, 16149 Genoa, Italy
| | - Michele Iafisco
- National Research Council (CNR), Institute of Science and Technology for Ceramics (ISTEC), Faenza, Italy
| | - Alessio Adamiano
- National Research Council (CNR), Institute of Science and Technology for Ceramics (ISTEC), Faenza, Italy
| | - Stefano Rossi
- CERT, Center of Excellence for Toxicological Research, Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Maurizio Aiello
- National Research Council (CNR), Institute of Electronic, Computer and Telecommunications (IEIIT), via de Marini 6, 16149 Genoa, Italy
| | - Maria Barandalla-Sobrados
- National Research Council (CNR), Institute of Genetic and Biomedical Research UOS Milan (IRGB), Milan, Italy
- Humanitas Clinical and Research Center, Rozzano, Milan, Italy
| | - Pierluigi Carullo
- National Research Council (CNR), Institute of Genetic and Biomedical Research UOS Milan (IRGB), Milan, Italy
- Humanitas Clinical and Research Center, Rozzano, Milan, Italy
| | - Michele Miragoli
- CERT, Center of Excellence for Toxicological Research, Department of Medicine and Surgery, University of Parma, Parma, Italy
- National Research Council (CNR), Institute of Genetic and Biomedical Research UOS Milan (IRGB), Milan, Italy
- Humanitas Clinical and Research Center, Rozzano, Milan, Italy
| | - Anna Tampieri
- National Research Council (CNR), Institute of Science and Technology for Ceramics (ISTEC), Faenza, Italy
| | - Silvia Scaglione
- National Research Council (CNR), Institute of Electronic, Computer and Telecommunications (IEIIT), via de Marini 6, 16149 Genoa, Italy
| | - Daniele Catalucci
- National Research Council (CNR), Institute of Genetic and Biomedical Research UOS Milan (IRGB), Milan, Italy
- Humanitas Clinical and Research Center, Rozzano, Milan, Italy
| |
Collapse
|
24
|
Yin Y, Chen P, Yu Q, Peng Y, Zhu Z, Tian J. The Effects of a Pulsed Electromagnetic Field on the Proliferation and Osteogenic Differentiation of Human Adipose-Derived Stem Cells. Med Sci Monit 2018; 24:3274-3282. [PMID: 29775452 PMCID: PMC5987610 DOI: 10.12659/msm.907815] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Background A low frequency pulsed electromagnetic field (PEMF) has been confirmed to play an important role in promoting the osteogenic differentiation of human bone marrow stem cells (BMSCs). Adipose-derived stem cells (ASCs) possess some attractive characteristics for clinical application compared to BMSCs, such as abundant stem cells from lipoaspirates, faster growth, less discomfort and morbidity during surgery. ASCs can become adipocytes, osteoblasts, chondrocytes, myocytes, neurocytes, and other cell types. Thus, ASCs might be a good alternative in clinical work involving treatment with PEMF. Material/Methods Human ASCs (hASCs)were divided into a control group (without PEMF exposure) and an experimental group (PEMF for two hours per day). We examined the effect of PEMF on promoting cell proliferation and osteogenic differentiation from several aspects: CCK-8 proliferation assay, RNA extraction, qRT-PCR detection, western blotting, and immunofluorescence staining experiments. Results PEMF could promote cell proliferation of human ASCs (hASCs) at an early stage as determined by CCK-8 assay. A specific intensity (1 mT) and frequency (50 Hz) of PEMF promoted osteogenic differentiation in hASCs in alkaline phosphatase (ALP) staining experiments. In addition, bone-related gene expression increased after two weeks of PEMF exposure, the protein expression of OPN, OCN, and RUNX-2 also increased after a longer period (three weeks) of PEMF treatment as determined by western blotting and immunofluorescence staining. Conclusions We found for the first time that PMEF has a role in stimulating cell proliferation of hASCs at an early period, subsequently promoting bone-related gene expression and inducing the expression of related proteins to stimulate osteogenic differentiation.
Collapse
Affiliation(s)
- Yukun Yin
- Department of Orthopedics, Zhujiang Hospital, Southern Medical University, Haizhu, Guangzhou, China (mainland)
| | - Ping Chen
- Department of Orthopedics, Zhujiang Hospital, Southern Medical University, Haizhu, Guangzhou, China (mainland)
| | - Qiang Yu
- Department of Orthopedics, Zhujiang Hospital, Southern Medical University, Haizhu, Guangzhou, China (mainland)
| | - Yan Peng
- Department of Human Anatomy, Basic Medical College, Southern Medical University, Baiyun, Guangzhou, China (mainland)
| | - ZeHao Zhu
- Department of Orthopedics, Zhujiang Hospital, Southern Medical University, Haizhu, Guangzhou, China (mainland)
| | - Jing Tian
- Department of Orthopedics, Zhujiang Hospital,Southern Medical University, Haizhu, Guangzhou, China (mainland)
| |
Collapse
|
25
|
Bacakova L, Zarubova J, Travnickova M, Musilkova J, Pajorova J, Slepicka P, Kasalkova NS, Svorcik V, Kolska Z, Motarjemi H, Molitor M. Stem cells: their source, potency and use in regenerative therapies with focus on adipose-derived stem cells - a review. Biotechnol Adv 2018; 36:1111-1126. [PMID: 29563048 DOI: 10.1016/j.biotechadv.2018.03.011] [Citation(s) in RCA: 323] [Impact Index Per Article: 53.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2018] [Revised: 03/12/2018] [Accepted: 03/15/2018] [Indexed: 02/08/2023]
Abstract
Stem cells can be defined as units of biological organization that are responsible for the development and the regeneration of organ and tissue systems. They are able to renew their populations and to differentiate into multiple cell lineages. Therefore, these cells have great potential in advanced tissue engineering and cell therapies. When seeded on synthetic or nature-derived scaffolds in vitro, stem cells can be differentiated towards the desired phenotype by an appropriate composition, by an appropriate architecture, and by appropriate physicochemical and mechanical properties of the scaffolds, particularly if the scaffold properties are combined with a suitable composition of cell culture media, and with suitable mechanical, electrical or magnetic stimulation. For cell therapy, stem cells can be injected directly into damaged tissues and organs in vivo. Since the regenerative effect of stem cells is based mainly on the autocrine production of growth factors, immunomodulators and other bioactive molecules stored in extracellular vesicles, these structures can be isolated and used instead of cells for a novel therapeutic approach called "stem cell-based cell-free therapy". There are four main sources of stem cells, i.e. embryonic tissues, fetal tissues, adult tissues and differentiated somatic cells after they have been genetically reprogrammed, which are referred to as induced pluripotent stem cells (iPSCs). Although adult stem cells have lower potency than the other three stem cell types, i.e. they are capable of differentiating into only a limited quantity of specific cell types, these cells are able to overcome the ethical and legal issues accompanying the application of embryonic and fetal stem cells and the mutational effects associated with iPSCs. Moreover, adult stem cells can be used in autogenous form. These cells are present in practically all tissues in the organism. However, adipose tissue seems to be the most advantageous tissue from which to isolate them, because of its abundancy, its subcutaneous location, and the need for less invasive techniques. Adipose tissue-derived stem cells (ASCs) are therefore considered highly promising in present-day regenerative medicine.
Collapse
Affiliation(s)
- Lucie Bacakova
- Department of Biomaterials and Tissue Engineering, Institute of Physiology of the Czech Academy of Sciences, Videnska 1083, 14220 Prague, 4-Krc, Czech Republic.
| | - Jana Zarubova
- Department of Biomaterials and Tissue Engineering, Institute of Physiology of the Czech Academy of Sciences, Videnska 1083, 14220 Prague, 4-Krc, Czech Republic
| | - Martina Travnickova
- Department of Biomaterials and Tissue Engineering, Institute of Physiology of the Czech Academy of Sciences, Videnska 1083, 14220 Prague, 4-Krc, Czech Republic
| | - Jana Musilkova
- Department of Biomaterials and Tissue Engineering, Institute of Physiology of the Czech Academy of Sciences, Videnska 1083, 14220 Prague, 4-Krc, Czech Republic
| | - Julia Pajorova
- Department of Biomaterials and Tissue Engineering, Institute of Physiology of the Czech Academy of Sciences, Videnska 1083, 14220 Prague, 4-Krc, Czech Republic
| | - Petr Slepicka
- Department of Solid State Engineering, University of Chemistry and Technology Prague, Technicka 5, 166 28 Prague, 6-Dejvice, Czech Republic
| | - Nikola Slepickova Kasalkova
- Department of Solid State Engineering, University of Chemistry and Technology Prague, Technicka 5, 166 28 Prague, 6-Dejvice, Czech Republic
| | - Vaclav Svorcik
- Department of Solid State Engineering, University of Chemistry and Technology Prague, Technicka 5, 166 28 Prague, 6-Dejvice, Czech Republic
| | - Zdenka Kolska
- Faculty of Science, J.E. Purkyne University, Ceske mladeze 8, 400 96 Usti nad Labem, Czech Republic
| | - Hooman Motarjemi
- Clinic of Plastic Surgery, Faculty Hospital Na Bulovce, Budinova 67/2, 180 81 Prague, 8-Liben, Czech Republic
| | - Martin Molitor
- Clinic of Plastic Surgery, Faculty Hospital Na Bulovce, Budinova 67/2, 180 81 Prague, 8-Liben, Czech Republic
| |
Collapse
|
26
|
Ma R, Liang J, Huang W, Guo L, Cai W, Wang L, Paul C, Yang HT, Kim HW, Wang Y. Electrical Stimulation Enhances Cardiac Differentiation of Human Induced Pluripotent Stem Cells for Myocardial Infarction Therapy. Antioxid Redox Signal 2018; 28:371-384. [PMID: 27903111 PMCID: PMC5770128 DOI: 10.1089/ars.2016.6766] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
AIMS Electrical stimulation (EleS) can promote cardiac differentiation, but the underlying mechanism is not well known. This study investigated the effect of EleS on cardiomyocyte (CM) differentiation of human induced pluripotent stem cells (hiPSCs) and evaluated the therapeutic effects for the treatment of myocardial infarction (MI). RESULTS Cardiac differentiation of hiPSCs was induced with EleS after embryoid body formation. Spontaneously beating hiPSCs were observed as early at 2 days when treated with EleS compared with control treatment. The cardiac differentiation efficiency of hiPSCs was significantly enhanced by EleS. In addition, the functional maturation of hiPSC-CMs under EleS was confirmed by calcium indicators, intracellular Ca2+ levels, and expression of structural genes. Mechanistically, EleS mediated cardiac differentiation of hiPSCs through activation of Ca2+/PKC/ERK pathways, as revealed by RNA sequencing, quantitative polymerase chain reaction, and Western blotting. After transplantation in immunodeficient MI mice, EleS-preconditioned hiPSC-derived cells significantly improved cardiac function and attenuated expansion of infarct size. The preconditioned hiPSC-derived CMs were functionally integrated with the host heart. INNOVATION We show EleS as an efficacious time-saving approach for CM generation. The global RNA profiling shows that EleS can accelerate cardiac differentiation of hiPSCs through activation of multiple pathways. The cardiac-mimetic electrical signals will provide a novel approach to generate functional CMs and facilitate cardiac tissue engineering for successful heart regeneration. CONCLUSION EleS can enhance efficiency of cardiac differentiation in hiPSCs and promote CM maturation. The EleS-preconditioned CMs emerge as a promising approach for clinical application in MI treatment. Antioxid. Redox Signal. 28, 371-384.
Collapse
Affiliation(s)
- Ruilian Ma
- 1 Department of Pathology and Laboratory Medicine, College of Medicine, University of Cincinnati , Cincinnati, Ohio
| | - Jialiang Liang
- 1 Department of Pathology and Laboratory Medicine, College of Medicine, University of Cincinnati , Cincinnati, Ohio
| | - Wei Huang
- 1 Department of Pathology and Laboratory Medicine, College of Medicine, University of Cincinnati , Cincinnati, Ohio
| | - Linlin Guo
- 1 Department of Pathology and Laboratory Medicine, College of Medicine, University of Cincinnati , Cincinnati, Ohio
| | - Wenfeng Cai
- 1 Department of Pathology and Laboratory Medicine, College of Medicine, University of Cincinnati , Cincinnati, Ohio
| | - Lei Wang
- 1 Department of Pathology and Laboratory Medicine, College of Medicine, University of Cincinnati , Cincinnati, Ohio
| | - Christian Paul
- 1 Department of Pathology and Laboratory Medicine, College of Medicine, University of Cincinnati , Cincinnati, Ohio
| | - Huang-Tian Yang
- 2 Key Laboratory of Stem Cell Biology and Laboratory of Molecular Cardiology, Institute of Health Sciences, Institutes for Biological Sciences (SIBS), Chinese Academy of Sciences (CAS) and Shanghai Jiao Tong University School of Medicine (SJTUSM) , Shanghai, China
| | - Ha Won Kim
- 1 Department of Pathology and Laboratory Medicine, College of Medicine, University of Cincinnati , Cincinnati, Ohio
| | - Yigang Wang
- 1 Department of Pathology and Laboratory Medicine, College of Medicine, University of Cincinnati , Cincinnati, Ohio
| |
Collapse
|
27
|
Hajipour Verdom B, Abdolmaleki P, Behmanesh M. The Static Magnetic Field Remotely Boosts the Efficiency of Doxorubicin through Modulating ROS Behaviors. Sci Rep 2018; 8:990. [PMID: 29343746 PMCID: PMC5772617 DOI: 10.1038/s41598-018-19247-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Accepted: 12/18/2017] [Indexed: 01/27/2023] Open
Abstract
Exposure to magnetic field (MF) can affect cellular metabolism remotely. Cardio-toxic effects of Doxorubicin (DOXO) have limited clinical uses at high dose. MF due to its effect on reactive oxygen species (ROS) lifetime, may provide a suitable choice to boost the efficacy of this drug at low dose. Here, we investigated the potential effects of homogenous static magnetic field (SMF) on DOXO-induced toxicity and proliferation rate of cancer cells. The results indicated that SMF similar to DOXO decreased the cell viability as well as the proliferation rate of MCF-7 and HFF cells. Moreover, combination of 10 mT SMF and 0.1 µM DOXO decreased the viability and proliferation rate of cancer and normal cells in a synergetic manner. In spite of high a GSH level in cancer cell, SMF boosts the generation and lifetime of ROS at low dose of DOXO, and overcame to GSH mediated drug resistance. The results also confirmed that SMF exposure decreased 50% iron content of cells, which is attributed to iron homeostasis. In conclusion, these findings suggest that SMF can decrease required dose of chemotherapy drugs such as DOXO and thereby decrease their side effect.
Collapse
Affiliation(s)
- Behnam Hajipour Verdom
- Department of Biophysics, Faculty of Biological Sciences, Tarbiat Modares University (TMU), Tehran, Iran
| | - Parviz Abdolmaleki
- Department of Biophysics, Faculty of Biological Sciences, Tarbiat Modares University (TMU), Tehran, Iran.
| | - Mehrdad Behmanesh
- Department of Genetics, Faculty of Biological Sciences, Tarbiat Modares University (TMU), Tehran, Iran
| |
Collapse
|
28
|
Magnetic Fields and Reactive Oxygen Species. Int J Mol Sci 2017; 18:ijms18102175. [PMID: 29057846 PMCID: PMC5666856 DOI: 10.3390/ijms18102175] [Citation(s) in RCA: 124] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Revised: 09/30/2017] [Accepted: 10/13/2017] [Indexed: 12/21/2022] Open
Abstract
Reactive oxygen species (ROS) ubiquitously exist in mammalian cells to participate in various cellular signaling pathways. The intracellular ROS levels are dependent on the dynamic balance between ROS generation and elimination. In this review, we summarize reported studies about the influences of magnetic fields (MFs) on ROS levels. Although in most cases, MFs increased ROS levels in human, mouse, rat cells, and tissues, there are also studies showing that ROS levels were decreased or not affected by MFs. Multiple factors could cause these discrepancies, including but not limited to MF type/intensity/frequency, exposure time and assay time-point, as well as different biological samples examined. It will be necessary to investigate the influences of different MFs on ROS in various biological samples systematically and mechanistically, which will be helpful for people to get a more complete understanding about MF-induced biological effects. In addition, reviewing the roles of MFs in ROS modulation may open up new scenarios of MF application, which could be further and more widely adopted into clinical applications, particularly in diseases that ROS have documented pathophysiological roles.
Collapse
|
29
|
Kolanowski TJ, Antos CL, Guan K. Making human cardiomyocytes up to date: Derivation, maturation state and perspectives. Int J Cardiol 2017; 241:379-386. [DOI: 10.1016/j.ijcard.2017.03.099] [Citation(s) in RCA: 75] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2017] [Revised: 02/24/2017] [Accepted: 03/21/2017] [Indexed: 12/29/2022]
|
30
|
Mao L, Wang H, Ma F, Guo Z, He H, Zhou H, Wang N. Exposure to static magnetic fields increases insulin secretion in rat INS-1 cells by activating the transcription of the insulin gene and up-regulating the expression of vesicle-secreted proteins. Int J Radiat Biol 2017; 93:831-840. [PMID: 28593826 DOI: 10.1080/09553002.2017.1332439] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
PURPOSE To evaluate the effect of static magnetic fields (SMFs) on insulin secretion and explore the mechanisms underlying exposure to SMF-induced insulin secretion in rat insulinoma INS-1 cells. MATERIALS AND METHODS INS-1 cells were exposed to a 400 mT SMF for 72 h, and the proliferation of INS-1 cells was detected by (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay. The secretion of insulin was measured with an enzyme linked immunosorbent assays (ELISA), the expression of genes was detected by real-time PCR, and the expression of proteins was measured by Western blotting. RESULTS Exposure to an SMF increased the expression and secretion of insulin by INS-1 cells but did not affect cell proliferation. Moreover, SMF exposure up-regulated the expression of several pancreas-specific transcriptional factors. Specifically, the activity of the rat insulin promoter was enhanced in INS-1 cells exposed to an SMF, and the expression levels of synaptosomal-associated protein 25 (SNAP-25) and syntaxin-1A were up-regulated after exposure to an SMF. CONCLUSIONS SMF exposure can promote insulin secretion in rat INS-1 cells by activating the transcription of the insulin gene and up-regulating the expression of vesicle-secreted proteins.
Collapse
Affiliation(s)
- Libin Mao
- a Key Laboratory of Industrial Fermentation Microbiology, Ministry of Education and Tianjin, College of Biotechnology , Tianjin University of Science and Technology , Tianjin , P.R. China
| | - Huiqin Wang
- a Key Laboratory of Industrial Fermentation Microbiology, Ministry of Education and Tianjin, College of Biotechnology , Tianjin University of Science and Technology , Tianjin , P.R. China
| | - Fenghui Ma
- a Key Laboratory of Industrial Fermentation Microbiology, Ministry of Education and Tianjin, College of Biotechnology , Tianjin University of Science and Technology , Tianjin , P.R. China
| | - Zhixia Guo
- a Key Laboratory of Industrial Fermentation Microbiology, Ministry of Education and Tianjin, College of Biotechnology , Tianjin University of Science and Technology , Tianjin , P.R. China
| | - Hongpeng He
- a Key Laboratory of Industrial Fermentation Microbiology, Ministry of Education and Tianjin, College of Biotechnology , Tianjin University of Science and Technology , Tianjin , P.R. China
| | - Hao Zhou
- a Key Laboratory of Industrial Fermentation Microbiology, Ministry of Education and Tianjin, College of Biotechnology , Tianjin University of Science and Technology , Tianjin , P.R. China
| | - Nan Wang
- a Key Laboratory of Industrial Fermentation Microbiology, Ministry of Education and Tianjin, College of Biotechnology , Tianjin University of Science and Technology , Tianjin , P.R. China
| |
Collapse
|
31
|
Hanini R, Chatti A, Ghorbel SB, Landoulsi A. Role of Sod Gene in Response to Static Magnetic Fields in Pseudomonas aeruginosa. Curr Microbiol 2017; 74:930-937. [PMID: 28523373 DOI: 10.1007/s00284-017-1264-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Accepted: 05/10/2017] [Indexed: 12/11/2022]
Abstract
The protective role of superoxide dismutase (SOD) against non-ionizing radiation such as static electromagnetic field (200 mT) has been studied in wild-type and mutant strain of Pseudomonas aeruginosa lacking cytosolic Mn-SOD (sodM), Fe-SOD (sodB), or both SODs (sodMB). Our results showed that inactivation of sodM and/or sodB genes increases the sensitivity of P. aeruginosa toward stress induced by the static magnetic field (200 mT). Furthermore, our results showed an enhancement of SOD, catalase, and peroxidases after exposure to the magnetic field. However, wild-type cells maintained significantly higher activities of antioxidant enzymes than mutant strains. The malondialdehyde produced by the oxidative degradation of unsaturated lipids and fatty acids showed significant increase in mutant strains compared to the wild-type. The overall results showed that the SOD has a protective role against a stress induced by static electromagnetic field in P. aeruginosa.
Collapse
Affiliation(s)
- Raouia Hanini
- Unité de Biochimie des lipides et interactions des macromolécules en Biologie (03/UR/0902), Laboratoire de Biochimie et biologie moléculaire, Faculté des Sciences de Bizerte, Zarzouna 7021, Bizerte, Tunisia.
| | - Abdelwaheb Chatti
- Laboratoire de Traitement des Eaux Usées, Centre de Recherches et Technologies des Eaux, Technopole Borj Cedria, Tunis, Tunisia
| | - Selma Ben Ghorbel
- Laboratoire de Traitement des Eaux Usées, Centre de Recherches et Technologies des Eaux, Technopole Borj Cedria, Tunis, Tunisia
| | - Ahmed Landoulsi
- Unité de Biochimie des lipides et interactions des macromolécules en Biologie (03/UR/0902), Laboratoire de Biochimie et biologie moléculaire, Faculté des Sciences de Bizerte, Zarzouna 7021, Bizerte, Tunisia
| |
Collapse
|
32
|
Effect of Extremely Low Frequency Electromagnetic Field on MAP2 and Nestin Gene Expression of Hair Follicle Dermal Papilla Cells. Int J Artif Organs 2016; 39:294-9. [PMID: 27515859 DOI: 10.5301/ijao.5000512] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/05/2016] [Indexed: 12/19/2022]
Abstract
Introduction In recent years, the extremely low frequency electromagnetic field (ELF-EMF) has attracted a great deal of scientific interest. The ELF-EMF signal is able to control ion transport across ion channels and therefore induce cell differentiation. Aim The purpose of this study was to investigate the effect of ELF-EMF (50 Hz, 1 mT) on MAP2 and Nestin gene expression of dermal papilla mesenchymal cells (DPCs). Methods In order to examine the effect of chemical and electromagnetic factors on gene expression, 4 experimental groups, namely chemical (cell exposure to chemical signals), EMF (exposing cells to ELF-EMF), chemical-EMF (subjecting cells to chemical signals and ELF-EMF) and control (with no treatment) groups, were prepared, treated for 5 days, and studied. To assess the effect of extended test time on the expression of neural differentiation markers (Nestin and MAP2), an EMF group was prepared and treated for a period of 14 consecutive days. The beneficial role of EMF in inducing neural differentiation was shown by real-time PCR analysis. Results The higher expression of MAP2 after 14 days compared to that after 5 days and decrease of cell proliferation on days 5 to 20 were indicative of the positive effect of extending treatment time on neural differentiation by evaluation of gene expression in EMF group.
Collapse
|
33
|
Bekhite MM, Finkensieper A, Abou-Zaid FA, El-Shourbagy IK, El-Fiky NK, Omar KM, Sauer H, Wartenberg M. Differential effects of high and low strength magnetic fields on mouse embryonic development and vasculogenesis of embryonic stem cells. Reprod Toxicol 2016; 65:46-58. [PMID: 27346840 DOI: 10.1016/j.reprotox.2016.06.016] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Revised: 06/01/2016] [Accepted: 06/22/2016] [Indexed: 01/24/2023]
Abstract
Man-made magnetic fields (MFs) may exert adverse effects on mammalian embryonic development. Herein, we analysed the effect of 10mT 50Hz sinusoidal (AC) or static (DC) MFs versus 1mT MFs on embryonic development of mice. Exposure for 20days during gestation to 10mT MFs increased resorptions and dead fetuses, decreased crown-rump length and fresh weight, reduced blood vessel differentiation and caused histological changes, accompanied with diminished vascular endothelial growth factor (VEGF) protein expression in several organs. In embryonic stem (ES) cell-derived embryoid bodies exposure towards 10mT MFs increased reactive oxygen species (ROS), decreased vascular marker as well as VEGF expression and enhanced apoptosis. In conclusion, our combined data from in vivo and in vitro experiments identified VEGF as an important mediator during embryonic development that can be influenced by high strength MFs, which in consequence leads to severe abnormalities in fetus organs and blood vessel formation.
Collapse
Affiliation(s)
- Mohamed M Bekhite
- University Heart Center, Clinic of Internal Medicine I, Department of Cardiology, Jena University Hospital, Jena, Germany; Department of Zoology, Faculty of Science, Tanta University, Tanta 31527, Egypt.
| | - Andreas Finkensieper
- University Heart Center, Clinic of Internal Medicine I, Department of Cardiology, Jena University Hospital, Jena, Germany
| | - Fouad A Abou-Zaid
- Department of Zoology, Faculty of Science, Tanta University, Tanta 31527, Egypt
| | | | - Nabil K El-Fiky
- Department of Zoology, Faculty of Science, Tanta University, Tanta 31527, Egypt
| | - Khaled M Omar
- Physics Department, Faculty of Science, Tanta University, 31527, Egypt
| | - Heinrich Sauer
- Department of Physiology, Justus Liebig University Giessen, Germany
| | - Maria Wartenberg
- University Heart Center, Clinic of Internal Medicine I, Department of Cardiology, Jena University Hospital, Jena, Germany
| |
Collapse
|
34
|
Albuquerque WWC, Costa RMPB, Fernandes TDSE, Porto ALF. Evidences of the static magnetic field influence on cellular systems. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2016; 121:16-28. [DOI: 10.1016/j.pbiomolbio.2016.03.003] [Citation(s) in RCA: 82] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/03/2015] [Accepted: 03/10/2016] [Indexed: 01/29/2023]
|
35
|
Safari M, Jadidi M, Baghian A, Hasanzadeh H. Proliferation and differentiation of rat bone marrow stem cells by 400μT electromagnetic field. Neurosci Lett 2015; 612:1-6. [PMID: 26639423 DOI: 10.1016/j.neulet.2015.11.044] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2015] [Revised: 11/25/2015] [Accepted: 11/25/2015] [Indexed: 10/22/2022]
Abstract
The interaction between environment electromagnetic field (EMF) and cells can effect on various physiological processes. EMF as an external inducing factor, could effect on proliferation or differentiation of cells. The purpose of this study was to evaluate the influence of the electromagnetic field on the viability, proliferation and differentiation rate of bone marrow stem cells (BMSCs) to neuron. BMSCs were obtained from 42 adult male rats. The cells incubated and cultured in 96-wells and 6-wells plates and exposed to electromagnetic field (40 or 400μT) with a selected waveform: AC (alternative current), rectified half wave (RHW) and rectified full wave (RFW), for a week. To assess the viability and proliferation rate of treated cells, MTT assay was done, and then immunocytochemistry staining Neu N was used to evaluate cell differentiation to neuron. Results showed that EMF decreases the viability and proliferation in treated groups. But in AC group's reduction was significant. Minimum viability and proliferation rate was observed in RHW 400μT group compared with sham. Immunocytochemistry showed that EMF can induce BMSC differentiation into neuron in AC 400μT and RFW 400μT. Evidences of this research support the hypothesis that EMF can induce differentiation of BMSCs to neuron.
Collapse
Affiliation(s)
- Manouchehr Safari
- Research Center of Nervous System Stem Cells and Department of Anatomy, School of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| | - Majid Jadidi
- Department of Medical Physics, School of Medicine, Semnan University of Medical Sciences, Semnan, Iran.
| | - Atefeh Baghian
- Department of Medical Physics, School of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| | - Hadi Hasanzadeh
- Department of Medical Physics, School of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| |
Collapse
|
36
|
Cardiomyogenesis of embryonic stem cells upon purinergic receptor activation by ADP and ATP. Purinergic Signal 2015; 11:491-506. [PMID: 26395809 DOI: 10.1007/s11302-015-9468-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2015] [Accepted: 09/09/2015] [Indexed: 02/07/2023] Open
Abstract
Purinergic signaling may be involved in embryonic development of the heart. In the present study, the effects of purinergic receptor stimulation on cardiomyogenesis of mouse embryonic stem (ES) cells were investigated. ADP or ATP increased the number of cardiac clusters and cardiac cells, as well as beating frequency. Cardiac-specific genes showed enhanced expression of α-MHC, MLC2v, α-actinin, connexin 45 (Cx45), and HCN4, on both gene and protein levels upon ADP/ATP treatment, indicating increased cardiomyogenesis and pacemaker cell differentiation. Real-time RT-PCR analysis of purinergic receptor expression demonstrated presence of P2X1, P2X4, P2X6, P2X7, P2Y1, P2Y2, P2Y4, and P2Y6 on differentiating ES cells. ATP and ADP as well as the P2X agonists β,γ-methylenadenosine 5'-triphosphate (β,γ-MetATP) and 8-bromoadenosine 5'-triphosphate (8-Br-ATP) but not UTP or UDP transiently increased the intracellular calcium concentration ([Ca(2+)](i)) as evaluated by the calcium indicator Fluo-4, whereas no changes in membrane potential were observed. [Ca(2+)](i) transients induced by ADP/ATP were abolished by the phospholipase C-β (PLC-β) inhibitor U-73122, suggesting involvement of metabotropic P2Y receptors. Furthermore, partial inhibition of [Ca(2+)](i) transients was achieved in presence of MRS2179, a selective P2Y1 receptor antagonist, whereas PPADS, a non-selective P2 receptor inhibitor, completely abolished the [Ca(2+)](i) response. Consequently, cardiomyocyte differentiation was decreased upon long term co-incubation of cells with ADP and P2 receptor antagonists. In summary, activation of purinoceptors and the subsequent [Ca(2+)](i) transients enhance the differentiation of ES cells toward cardiomyocytes. Purinergic receptor stimulation may be a promising strategy to drive the fate of pluripotent ES cells into a particular population of cardiomyocytes.
Collapse
|
37
|
Zhang K, Chen W, Bu T, Qi H, Sun R, He X. Decreased P-glycoprotein is associated with the inhibitory effects of static magnetic fields and cisplatin on K562 cells. Bioelectromagnetics 2014; 35:437-43. [PMID: 25099308 DOI: 10.1002/bem.21863] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2013] [Accepted: 03/23/2014] [Indexed: 11/06/2022]
Abstract
In this study, we explored the mechanism of the killing effects of a moderate-intensity static magnetic field (SMF) and cisplatin (DDP) on K562 cells. We analyzed the metabolic activity of cells, the extracellular DDP content, and P-glycoprotein (P-gp) expression after K562 cells were exposed continuously to a uniform 8.8 mT SMF for 8 h, with or without DDP. We found that SMF combined with DDP (10 µg/ml) significantly inhibited the metabolic activity of K562 cells (P < 0.05), while neither DDP nor SMF alone affected the metabolic activity of these cells. In the SMF + DDP group, extracellular DDP content was significantly reduced (P < 0.05). DDP also induced the expression of P-gp (P < 0.05). By contrast, in the SMF + DDP group, P-gp expression decreased compared with the DDP group (P < 0.05). Taken together, our results showed that 8.8 mT SMF enhanced the killing potency of DDP on K562 cells by decreasing the expression of P-gp.
Collapse
Affiliation(s)
- Kun Zhang
- College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi, P. R. China
| | | | | | | | | | | |
Collapse
|
38
|
An in vitro study of the impact of 4mT static magnetic field to modify the differentiation rate of rat bone marrow stem cells into primordial germ cells. Differentiation 2014; 87:230-7. [PMID: 25037498 DOI: 10.1016/j.diff.2014.06.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2014] [Revised: 06/13/2014] [Accepted: 06/18/2014] [Indexed: 02/06/2023]
Abstract
This investigation was performed to evaluate the differentiation capacity and alteration in genes expression patterns during in vitro differentiation of bone marrow stem cells into primordial germ cells using static magnetic field (4mT) and BMP-4 (25ng/ml). The rate of differentiation was investigated using the Real Time-PCR method for tracing expression of differentiation markers (Oct-4, Nanog, C-Myc, Fragilis, Mvh and Stella). Then, immunocytochemical reaction was carried out for detection of marker proteins (Oct4 and Mvh). Increasing of the exposure time of the 4mT SMF (24 and 48h) and treatment time with 25ng/ml BMP4 (48 and 96h) indicated a marked decrease in expression of pluripotency genes (Oct-4, Nanog and C-Myc) and Oct4 protein and increase in primordial germ cell-specific genes (Fragilis, Mvh and Stella) and Mvh protein compared with the control group. Final results showed that in a synergistic manner, the combination of SMF with BMP4 exaggerates the differentiation potential of BMSCs to PGCs by activating the MAPK pathway and altering the Ca(2+) concentration.
Collapse
|
39
|
Razavi S, Salimi M, Shahbazi-Gahrouei D, Karbasi S, Kermani S. Extremely low-frequency electromagnetic field influences the survival and proliferation effect of human adipose derived stem cells. Adv Biomed Res 2014; 3:25. [PMID: 24592372 PMCID: PMC3928843 DOI: 10.4103/2277-9175.124668] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2013] [Accepted: 05/05/2013] [Indexed: 11/04/2022] Open
Abstract
BACKGROUND Extremely low-frequency electromagnetic fields (ELF-EMF) can effect on biological systems and alters some cell functions like proliferation rate. Therefore, we aimed to attempt the evaluation effect of ELF-EMF on the growth of human adipose derived stem cells (hADSCs). MATERIALS AND METHODS ELF-EMF was generated by a system including autotransformer, multi-meter, solenoid coils, teslameter and its probe. We assessed the effect of ELF-EMF with intensity of 0.5 and 1 mT and power line frequency 50 Hz on the survival of hADSCs for 20 and 40 min/day for 7 days by MTT assay. One-way analysis of variance was used to assessment the significant differences in groups. RESULTS ELF-EMF has maximum effect with intensity of 1 mT for 20 min/day on proliferation of hADSCs. The survival and proliferation effect (PE) in all exposure groups were significantly higher than that in sham groups (P < 0.05) except in group of 1 mT and 40 min/day. CONCLUSION Our results show that between 0.5 m and 1 mT ELF-EMF could be enhances survival and PE of hADSCs conserving the duration of exposure.
Collapse
Affiliation(s)
- Shahnaz Razavi
- Department of Anatomical Sciences, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Marzieh Salimi
- Department of Medical Physics and Medical Engineering, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Daryoush Shahbazi-Gahrouei
- Department of Medical Physics and Medical Engineering, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Saeed Karbasi
- Department of Medical Physics and Medical Engineering, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Saeed Kermani
- Department of Medical Physics and Medical Engineering, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|