1
|
Zhang L, Ma M, Li J, Qiao K, Xie Y, Zheng Y. Stimuli-responsive microcarriers and their application in tissue repair: A review of magnetic and electroactive microcarrier. Bioact Mater 2024; 39:147-162. [PMID: 38808158 PMCID: PMC11130597 DOI: 10.1016/j.bioactmat.2024.05.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 04/07/2024] [Accepted: 05/07/2024] [Indexed: 05/30/2024] Open
Abstract
Microcarrier applications have made great advances in tissue engineering in recent years, which can load cells, drugs, and bioactive factors. These microcarriers can be minimally injected into the defect to help reconstruct a good microenvironment for tissue repair. In order to achieve more ideal performance and face more complex tissue damage, an increasing amount of effort has been focused on microcarriers that can actively respond to external stimuli. These microcarriers have the functions of directional movement, targeted enrichment, material release control, and providing signals conducive to tissue repair. Given the high controllability and designability of magnetic and electroactive microcarriers, the research progress of these microcarriers is highlighted in this review. Their structure, function and applications, potential tissue repair mechanisms, and challenges are discussed. In summary, through the design with clinical translation ability, meaningful and comprehensive experimental characterization, and in-depth study and application of tissue repair mechanisms, stimuli-responsive microcarriers have great potential in tissue repair.
Collapse
Affiliation(s)
- LiYang Zhang
- School of Material Science and Engineering, University of Science and Technology Beijing, Beijing, China
| | - Mengjiao Ma
- Beijing Wanjie Medical Device Co., Ltd, Beijing, China
| | - Junfei Li
- School of Material Science and Engineering, University of Science and Technology Beijing, Beijing, China
| | - Kun Qiao
- Beijing Gerecov Technology Company Ltd., Beijing, China
| | - Yajie Xie
- Beijing Gerecov Technology Company Ltd., Beijing, China
| | - Yudong Zheng
- School of Material Science and Engineering, University of Science and Technology Beijing, Beijing, China
| |
Collapse
|
2
|
Jaiswal J, Dhayal M. Rapid neurogenic differentiation of human mesenchymal stem cells through electrochemical stimulation. Bioelectrochemistry 2023; 153:108468. [PMID: 37224602 DOI: 10.1016/j.bioelechem.2023.108468] [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: 09/17/2022] [Revised: 05/17/2023] [Accepted: 05/18/2023] [Indexed: 05/26/2023]
Abstract
The neurogenic differentiation of human mesenchymal stem cells (hMSCs) has been substantially handicapped with the choice of chemical or electrical stimulations for long durations. We demonstrate an innovative strategy of stimulation with <1.0 V for <200 s to achieve hMSCs differentiation towards neural progenitor cells within 24 h and their commitment towards differentiation to neurons on day 3 with the use of three-electrode electrostimulation. Stimulated hMSCs (ES hMSCs) showed elevated expression of neural-specific markers and mitochondrial membrane potential. A voltage bias of ±0.5 V and ±1.0 V did not show any adverse effect on cell viability and proliferation, whereas cells stimulated with ±1.5 V showed an upsurge in the dead cell populations. With the progression of time after stimulation, a rise in mitochondrial membrane potential (MMP, ΔΨ M) was observed in the ES hMSCs and thereby generating intracellular reactive oxygen species (ROS), acting as a key messenger to induce neuronal differentiation. The stratagem may provide insightful handles to circumvent neurodifferentiation impediments, a focal issue for regenerative medicine.
Collapse
Affiliation(s)
- Juhi Jaiswal
- Department of Physics, Indian Institute of Technology Delhi, Hauz Khas, New Delhi 110016, India; Nano-Cellular Medicine and Biophysics Laboratory, School of Biomedical Engineering, Indian Institute of Technology (BHU), Varanasi 221005, Uttar Pradesh, India
| | - Marshal Dhayal
- Department of Physics, Indian Institute of Technology Delhi, Hauz Khas, New Delhi 110016, India; Nano-Cellular Medicine and Biophysics Laboratory, School of Biomedical Engineering, Indian Institute of Technology (BHU), Varanasi 221005, Uttar Pradesh, India.
| |
Collapse
|
3
|
Safrole oxide induced 5-HT neuron-like cell differentiation of bone marrow mesenchymal stem cells by elevating G9a. In Vitro Cell Dev Biol Anim 2022; 58:513-520. [PMID: 35913528 DOI: 10.1007/s11626-021-00594-2] [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: 01/29/2021] [Accepted: 05/17/2021] [Indexed: 11/05/2022]
Abstract
In our previous study, we found that safrole oxide (SFO) could induce bone marrow mesenchymal stem cell differentiation into neuron-like cells. However, which kind of neuron cells was induced by SFO was unknown. Here, we found that SFO could induce BMSC differentiation into 5-hydroxytryptamine (5-HT) neuron-like cells. Microarray analysis of BMSCs treated with SFO for 6 h revealed a total of 35 genes changed more than twice. We selected G9a, a histone methyltransferase for further study. The upregulation of G9a was confirmed by RT-PCR and Western blot analysis. Small interfering RNA knockdown of G9a blocked SFO-induced BMSC differentiation. These results demonstrated that G9a was the pivotal factor in SFO-medicated 5-HT neuronal differentiation of BMSCs. Our findings provide a new clue for further investigating the gene control of BMSC differentiation into 5-HT neuron-like cells and provide a putative strategy for depression diseases therapies.
Collapse
|
4
|
Zeng Z, Yang Y, Deng J, Saif Ur Rahman M, Sun C, Xu S. Physical Stimulation Combined with Biomaterials Promotes Peripheral Nerve Injury Repair. Bioengineering (Basel) 2022; 9:292. [PMID: 35877343 PMCID: PMC9311987 DOI: 10.3390/bioengineering9070292] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 06/11/2022] [Accepted: 06/13/2022] [Indexed: 11/17/2022] Open
Abstract
Peripheral nerve injury (PNI) is a clinical problem with high morbidity that can cause severe damage. Surgical suturing or implants are usually required due to the slow speed and numerous factors affecting repair after PNI. An autologous nerve graft is the gold standard for PNI repair among implants. However, there is a potential problem of the functional loss of the donor site. Therefore, tissue-engineered nerve biomaterials are often used to bridge the gap between nerve defects, but the therapeutic effect is insufficient. In order to enhance the repair effect of nerve biomaterials for PNI, researchers are seeking to combine various stimulation elements, such as the addition of biological factors such as nerve growth factors or physical factors such as internal microstructural modifications of catheters and their combined application with physical stimulation therapy. Physical stimulation therapy is safer, is more convenient, and has more practical features than other additive factors. Its feasibility and convenience, when combined with nerve biomaterials, provide broader application prospects for PNI repair, and has therefore become a research hot spot. This paper will review the combined application of physical stimulation and biomaterials in PNI repair in recent years to provide new therapeutic ideas for the future use of physical stimulation in PNI repair.
Collapse
Affiliation(s)
- Zhipeng Zeng
- Institute for Advanced Study, Shenzhen University, Shenzhen 518060, China; (Z.Z.); (M.S.U.R.)
- Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, College of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen 518060, China
- School of Sport Medicine and Rehabilitation, Beijing Sport University, Beijing 100084, China
| | - Yajing Yang
- National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital and Shenzhen Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Shenzhen 518116, China;
| | - Junyong Deng
- Department of Rehabilitation, Second Affiliated Hospital of Nanchang University, Nanchang 330006, China;
| | - Muhammad Saif Ur Rahman
- Institute for Advanced Study, Shenzhen University, Shenzhen 518060, China; (Z.Z.); (M.S.U.R.)
- Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, College of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen 518060, China
| | - Chengmei Sun
- Institute for Advanced Study, Shenzhen University, Shenzhen 518060, China; (Z.Z.); (M.S.U.R.)
- Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, College of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen 518060, China
| | - Shanshan Xu
- Institute for Advanced Study, Shenzhen University, Shenzhen 518060, China; (Z.Z.); (M.S.U.R.)
- Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, College of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen 518060, China
| |
Collapse
|
5
|
Jadidi T, Asadian N, Jadidi M, Ali Vafaei A. EMF promote BMSCs differentiation and functional recovery in hemiparkinsonian rats. Neurosci Lett 2022; 784:136765. [PMID: 35777611 DOI: 10.1016/j.neulet.2022.136765] [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: 07/15/2021] [Revised: 06/25/2022] [Accepted: 06/26/2022] [Indexed: 10/17/2022]
Abstract
Bone marrow mesenchymal stem cells (BMSCs) have self-renewal ability while maintaining the proliferation facility. The BMSCs reproducing ability could affect by electromagnetic fields (EMFs) as a physical inducing factor. We focused on the EMF (400 µT, 75 Hz) exposed multi-potential BMSCs which differentiated and successfully implanted in the substantia nigra pars compacta (SNpc) of Parkinson's disease rat model. The purified BMSCs are exposed to sinusoidal and square waveform EMF (1h/1 week or 7h/1 day) then injected into the left SNpc of Parkinson's rats. To evaluate the morphology of EMF exposed BMSCs, the cresyl violet staining labeled the Nissl bodies. After evaluation of the rat's activity by behavioral tests (open-field and rotarod tests), the brains were obtained for the preparation of SNpc blocks and carry out the cresyl violet staining. Cell morphology proved most cell differentiation to neurons in the sinusoidal EMF groups. In the sinusoidal EMF exposure groups, large and small neurons were seen with apparent synapses. Although in the square EMF exposed groups some neurons were seen, most of the differentiated cells were astrocytes, microglia, and oligodendrocyte. The results confirmed an improvement in locomotors' activity of BMSC alone and sinusoidal EMF exposed groups. We presented a low-frequency EMF (75 Hz) to promote the capability of BMSC proliferation, differentiation to neurons and glial cells, and motor coordination activity in the treatment of hemiparkinsonian rats.
Collapse
Affiliation(s)
- Taha Jadidi
- Nervous System Stem Cells Research Center, Semnan University of Medical Sciences, Semnan, Iran.
| | - Nader Asadian
- Department of Medical Physics, Semnan University of Medical Sciences, Semnan, Iran
| | - Majid Jadidi
- Department of Medical Physics, Semnan University of Medical Sciences, Semnan, Iran
| | - Abbas Ali Vafaei
- Physiology Research Center, Semnan University of Medical Sciences, Semnan, Iran
| |
Collapse
|
6
|
Lai H. Neurological effects of static and extremely-low frequency electromagnetic fields. Electromagn Biol Med 2022; 41:201-221. [DOI: 10.1080/15368378.2022.2064489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Affiliation(s)
- Henry Lai
- Department of Bioengineering, University of Washington, Seattle, WA, USA
| |
Collapse
|
7
|
Jaiswal J, Dhayal M. Electrochemically differentiated human MSCs biosensing platform for quantification of nestin and β-III tubulin as whole-cell system. Biosens Bioelectron 2022; 206:114134. [PMID: 35276463 DOI: 10.1016/j.bios.2022.114134] [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: 12/16/2021] [Revised: 02/09/2022] [Accepted: 02/23/2022] [Indexed: 12/14/2022]
Abstract
Polydimethylsiloxane (PDMS) on ITO substrate was used to create a well with conducting surface to adhere human mesenchymal stem cells (hMSCs) and provide electrochemical stimulation for inducing their differentiation into neural-like cells. The cells that received electrochemical stimulation did not show any noticeable change in their viability and proliferation. The cell morphology of the differentiated hMSCs adherent on ITO showed outgrowth and elongation in one dimension, resembling neural-like cells. Immunocytochemistry assessment by quantifying the expression of nestin and β-III tubulin also confirmed the differentiation of hMSCs. These differentiated hMSCs adherent on ITO were used as electrochemical biosensing platform for differential pulse voltammetry (DPV) measurement for selectively quantifying cell surface markers expressed by neural stem cells and mature neurons. The variation of nestin antibodies concentrations from 9 μU to 27 μU showed a linear increase in DPV current with a detection sensitivity of ∼28 nA/μU of antibody. Varying concentrations of β-III tubulin antibodies from 30 μU to 210 μU showed a linear increase in DPV current with a detection sensitivity of ∼2.0 nA/μU of antibody. The highest expression level of cell surface marker corresponding to β-III tubulin in total adherent cells on ITO was calculated. It was in the order of 10-8 U of antibodies/cell, representing the total population of mature neuron cells. This new way of detection may rapidly assess the quantitative expression of cell surface markers/antigens.
Collapse
Affiliation(s)
- Juhi Jaiswal
- Nano-Cellular Medicine and Biophysics Laboratory, School of Biomedical Engineering, Indian Institute of Technology (BHU), Varanasi, 221005, Uttar Pradesh, India
| | - Marshal Dhayal
- Nano-Cellular Medicine and Biophysics Laboratory, School of Biomedical Engineering, Indian Institute of Technology (BHU), Varanasi, 221005, Uttar Pradesh, India.
| |
Collapse
|
8
|
DNMT1 and miRNAs: possible epigenetics footprints in electromagnetic fields utilization in oncology. Med Oncol 2021; 38:125. [PMID: 34495398 DOI: 10.1007/s12032-021-01574-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Accepted: 09/01/2021] [Indexed: 10/20/2022]
Abstract
Many studies were performed to unravel the effects of different types of Electromagnetic fields (EMFs) on biological systems. Some studies were conducted to exploit EMFs for medical purposes mainly in cancer therapy. Although many studies suggest that the EMFs exposures can be effective in pre-clinical cancer issues, the treatment outcomes of these exposures on the cancer cells, especially at the molecular level, are challenging and overwhelmingly complicated yet. This article aims to review the epigenetic mechanisms that can be altered by EMFs exposures with the main emphasis on Extremely low frequency electromagnetic field (ELF-EMF). The epigenetic mechanisms are reversible and affected by environmental factors, thus, EMFs exposures can modulate these mechanisms. According to the reports, ELF-EMF exposures affect epigenetic machinery directly or through the molecular signaling pathways. ELF-EMF in association with DNA methylation, histone modification, miRNAs, and nucleosome remodeling could affect the homeostasis of cancer cells and play a role in DNA damage repairing, apoptosis induction, prevention of metastasis, differentiation, and cell cycle regulation. In general, the result of this study shows that ELF-EMF exposure probably can be effective in cancer epigenetic therapy, but more molecular and clinical investigations are needed to clarify the safe and specific dosimetric characteristics of ELF-EMF in practice.
Collapse
|
9
|
Gao Q, Leung A, Yang YH, Lau BWM, Wang Q, Liao LY, Xie YJ, He CQ. Extremely low frequency electromagnetic fields promote cognitive function and hippocampal neurogenesis of rats with cerebral ischemia. Neural Regen Res 2021; 16:1252-1257. [PMID: 33318402 PMCID: PMC8284293 DOI: 10.4103/1673-5374.301020] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Extremely low frequency electromagnetic fields (ELF-EMF) can improve the learning and memory impairment of rats with Alzheimer’s disease, however, its effect on cerebral ischemia remains poorly understood. In this study, we established rat models of middle cerebral artery occlusion/reperfusion. One day after modeling, a group of rats were treated with ELF-EMF (50 Hz, 1 mT) for 2 hours daily on 28 successive days. Our results showed that rats treated with ELF-EMF required shorter swimming distances and latencies in the Morris water maze test than those of untreated rats. The number of times the platform was crossed and the time spent in the target quadrant were greater than those of untreated rats. The number of BrdU+ /NeuN+ cells, representing newly born neurons, in the hippocampal subgranular zone increased more in the treated than in untreated rats. Up-regulation in the expressions of Notch1, Hes1, and Hes5 proteins, which are the key factors of the Notch signaling pathway, was greatest in the treated rats. These findings suggest that ELF-EMF can enhance hippocampal neurogenesis of rats with cerebral ischemia, possibly by affecting the Notch signaling pathway. The study was approved by the Institutional Ethics Committee of Sichuan University, China (approval No. 2019255A) on March 5, 2019.
Collapse
Affiliation(s)
- Qiang Gao
- Department of Rehabilitation Medicine, West China Hospital of Sichuan University; Institute of Disaster Management and Reconstruction, Sichuan University-The Hong Kong Polytechnic University, Chengdu, Sichuan Province; Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hong Kong Special Administrative Region, China
| | - Aaron Leung
- Institute of Disaster Management and Reconstruction, Sichuan University-The Hong Kong Polytechnic University, Chengdu, Sichuan Province; Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hong Kong Special Administrative Region, China
| | - Yong-Hong Yang
- Department of Rehabilitation Medicine, West China Hospital of Sichuan University; Institute of Disaster Management and Reconstruction, Sichuan University-The Hong Kong Polytechnic University, Chengdu, Sichuan Province; Department of Rehabilitation Sciences, The Hong Kong Polytechnic University, Hong Kong Special Administrative Region, China
| | - Benson Wui-Man Lau
- Department of Rehabilitation Sciences, The Hong Kong Polytechnic University, Hong Kong Special Administrative Region, China
| | - Qian Wang
- Department of Rehabilitation Medicine, West China Hospital of Sichuan University; Institute of Disaster Management and Reconstruction, Sichuan University-The Hong Kong Polytechnic University, Chengdu, Sichuan Province, China
| | - Ling-Yi Liao
- Department of Rehabilitation Medicine, West China Hospital of Sichuan University, Chengdu, Sichuan Province, China
| | - Yun-Juan Xie
- Department of Rehabilitation Medicine, West China Hospital of Sichuan University, Chengdu, Sichuan Province, China
| | - Cheng-Qi He
- Department of Rehabilitation Medicine, West China Hospital of Sichuan University; Institute of Disaster Management and Reconstruction, Sichuan University-The Hong Kong Polytechnic University, Chengdu, Sichuan Province, China
| |
Collapse
|
10
|
Asadian N, Jadidi M, Safari M, Jadidi T, Gholami M. EMF frequency dependent differentiation of rat bone marrow mesenchymal stem cells to astrocyte cells. Neurosci Lett 2020; 744:135587. [PMID: 33373676 DOI: 10.1016/j.neulet.2020.135587] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2020] [Revised: 11/23/2020] [Accepted: 12/16/2020] [Indexed: 10/22/2022]
Abstract
The numerous factors regulate the bone marrow mesenchymal stem cell (BMMSC) self-renewal and differentiation response. We aimed to analyze the influence of electromagnetic field (EMF) as an external inducing factor on rat BMMSC differentiation and proliferation to neuron and astrocyte cells. BMMSCs extracted from the rats femurs and tibias and incubated in a cell-cultured CO2 incubator. After the third passages, the plates selected randomly and then divided into seven groups (Sham exposed, three groups of square, and three groups of sinusoidal waveform EMF (25, 50, and 75 Hz, 400 μT, 1 h/day). The BMMSCs exposed to EMF at the middle of a Helmholtz coil for 7 days. The viable cell counting and proliferation performed by the MTT test and BMMSC differentiation into the neuron and the astrocyte cell was studied by immunocytochemistry staining. The results confirmed BMMSC viability and proliferation rate reduction in sinusoidal 25 Hz, square 50 Hz and sinusoidal 75 Hz EMF groups compare to sham. The maximum BMMSC differentiation to neuron was considered in sinusoidal 50 Hz and 75 Hz EMF groups. The increase of BMMSC differentiation to astrocyte cell was frequency dependent and the most differentiation was shown in square 75 Hz, and sinusoidal 75 Hz EMF groups. In conclusion, the results suggest that both square and sinusoidal EMF could affect BMMSC development and differentiation to neuron and astrocyte cells. Further studies for the consequence of EMF with wider flux density and frequency on BMMSC are recommended.
Collapse
Affiliation(s)
- Nader Asadian
- Department of Medical Physics, Semnan University of Medical Sciences, Semnan, Iran
| | - Majid Jadidi
- Department of Medical Physics, Semnan University of Medical Sciences, Semnan, Iran.
| | - Manouchehr Safari
- Nervous System Stem Cells Research Center, Semnan University of Medical Sciences, Semnan, Iran
| | - Taha Jadidi
- Nervous System Stem Cells Research Center, Semnan University of Medical Sciences, Semnan, Iran
| | - Mahbobeh Gholami
- Department of Biochemistry, Semnan University of Medical Sciences, Semnan, Iran
| |
Collapse
|
11
|
Zuo H, Liu X, Li Y, Wang D, Hao Y, Yu C, Xu X, Peng R, Song T. The mitochondria/caspase-dependent apoptotic pathway plays a role in the positive effects of a power frequency electromagnetic field on Alzheimer's disease neuronal model. J Chem Neuroanat 2020; 109:101857. [PMID: 32918997 DOI: 10.1016/j.jchemneu.2020.101857] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 08/20/2020] [Accepted: 09/06/2020] [Indexed: 11/16/2022]
Abstract
In this study, rat pheochromocytoma (PC12) cells were induced into an Alzheimer's Disease (AD) neuronal model using nerve growth factor (NGF; 50 ng/mL) and Amyloid β25-35 (20 μmol/L). Changes in the morphological structure, cell viability, apoptosis rate, and expression of apoptosis-related protein induced by exposure to a power frequency electromagnetic field (PF-MF; 50 Hz, 100 μT, 24 h) were detected respectively by light and electron microscopy, the MTT assay, immunohistochemistry, flow cytometry and enzyme-linked immunosorbent assays. The results showed that 3-12 h after PF-MF exposure, the pathological injury was improved partly; metabolic activity was promoted and cell apoptosis was inhibited in the AD neuronal model. In addition, PF-MF exposure significantly inhibited the expression of Caspase8, Caspase3, and CytC, but increased the Bcl-2/Bax ratio of the AD neuronal model. Meanwhile, PF-MF seemed to have no effect on the expression of Fas and TNFR1. This study indicated that the mitochondria/caspase-dependent apoptotic pathway plays an important role in the positive effects of PF-MF on an AD neuronal model. The results suggested that PF-MF exposure might have potential therapeutic value for AD, and the underling molecular mechanisms still need further studies.
Collapse
Affiliation(s)
- Hongyan Zuo
- Department of Experimental Pathology, Beijing Institute of Radiation Medicine, Beijing 100850, China.
| | - Xiao Liu
- Department of Experimental Pathology, Beijing Institute of Radiation Medicine, Beijing 100850, China; Department ofPathology, Hainan Hospital of PLA General Hospital, Sanya 572013, China
| | - Yang Li
- Department of Experimental Pathology, Beijing Institute of Radiation Medicine, Beijing 100850, China; Anhui Medical University, Hefei 230032, China
| | - Dewen Wang
- Department of Experimental Pathology, Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Yanhui Hao
- Department of Experimental Pathology, Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Chao Yu
- Department of Experimental Pathology, Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Xinping Xu
- Department of Experimental Pathology, Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Ruiyun Peng
- Department of Experimental Pathology, Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Tao Song
- Beijing Key Laboratory of Bioelectromagnetics, Institute of Electrical Engineering, Chinese Academy of Sciences, Beijing 100190, China
| |
Collapse
|
12
|
Yuan FY, Zhang MX, Shi YH, Li MH, Ou JY, Bai WF, Zhang MS. Bone marrow stromal cells-derived exosomes target DAB2IP to induce microglial cell autophagy, a new strategy for neural stem cell transplantation in brain injury. Exp Ther Med 2020; 20:2752-2764. [PMID: 32765770 PMCID: PMC7401953 DOI: 10.3892/etm.2020.9008] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Accepted: 05/13/2020] [Indexed: 02/06/2023] Open
Abstract
Bone marrow stromal cells (MSCs) are a useful source of stem cells for the treatment of various brain injury diseases due to their abundant supply and fewer ethical problems compared with transplant treatment. However, the clinical application of MSCs is limited due to allograft rejection and immunosuppression in the process of MSCs transplantation. According to previous studies, microglial cell autophagy occurs following co-culture with MSCs. In the present study, exosomes were obtained from MSCs and subsequently characterized using transmission electron microscopy, atomic force microscopy and dynamic light scattering particle size analysis. The type of microRNAs (miRs) found in the exosomes was then analyzed via gene chip. The results demonstrated that microglial cell autophagy could be induced by exosomes. This mechanism was therefore investigated further via reverse transcription-quantitative PCR, western blotting and luciferase assays. These results demonstrated that exosomes from MSCs could induce microglial cell autophagy through the miR-32-mediated regulation of disabled homolog 2-interacting protein, thus providing a theoretical basis for the clinical application of miRs in MSCs.
Collapse
Affiliation(s)
- Feng-Ying Yuan
- Department of Rehabilitation Medicine, The First Affiliated Hospital, Jinan University, Guangzhou, Guangdong 510632, P.R. China.,Department of Rehabilitation Medicine The First Affiliated Hospital, Guangdong Pharmaceutical University, Guangzhou, Guangdong 510600, P.R. China
| | - Ming-Xing Zhang
- Department of Rehabilitation Medicine The First Affiliated Hospital, Guangdong Pharmaceutical University, Guangzhou, Guangdong 510600, P.R. China
| | - Yi-Hua Shi
- Department of Rehabilitation Medicine The First Affiliated Hospital, Guangdong Pharmaceutical University, Guangzhou, Guangdong 510600, P.R. China
| | - Mei-Hui Li
- Department of Rehabilitation Medicine, Guangdong Provincial People's Hospital, Guangzhou, Guangdong 510120, P.R. China
| | - Jia-Yuan Ou
- Department of Rehabilitation Medicine, Guangdong Provincial People's Hospital, Guangzhou, Guangdong 510120, P.R. China
| | - Wen-Fang Bai
- Department of Rehabilitation Medicine, Guangdong Provincial People's Hospital, Guangzhou, Guangdong 510120, P.R. China.,Academy of Medical Sciences, Guangdong Provincial Institute of Geriatrics, Guangzhou, Guangdong 510080, P.R. China
| | - Ming-Sheng Zhang
- Department of Rehabilitation Medicine, The First Affiliated Hospital, Jinan University, Guangzhou, Guangdong 510632, P.R. China.,Department of Rehabilitation Medicine, Guangdong Provincial People's Hospital, Guangzhou, Guangdong 510120, P.R. China
| |
Collapse
|
13
|
Xia B, Gao J, Li S, Huang L, Zhu L, Ma T, Zhao L, Yang Y, Luo K, Shi X, Mei L, Zhang H, Zheng Y, Lu L, Luo Z, Huang J. Mechanical stimulation of Schwann cells promote peripheral nerve regeneration via extracellular vesicle-mediated transfer of microRNA 23b-3p. Theranostics 2020; 10:8974-8995. [PMID: 32802175 PMCID: PMC7415818 DOI: 10.7150/thno.44912] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Accepted: 06/09/2020] [Indexed: 12/14/2022] Open
Abstract
Rationale: Peripheral nerves are unique in their remarkable elasticity. Schwann cells (SCs), important components of the peripheral nervous system (PNS), are constantly subjected to physiological and mechanical stresses from dynamic stretching and compression forces during movement. So far, it is not clear if SCs sense and respond to mechanical signals. It is also unknown whether mechanical stimuli can interfere with the intercellular communications between neurons and SCs, and what role extracellular vesicles (EVs) play in this process. The present study aimed to examine the effect of mechanical stimuli on the EV-mediated intercellular communication between neurons and SCs, explore their effect on axonal regeneration, and investigate the underlying mechanism. Methods: Purified SCs were stimulated using a magnetic force-based mechanical stimulation (MS) system and EVs were purified from mechanically stimulated SCs (MS-SCs-EVs) and non-stimulated SCs (SCs-EVs). The effect of MS-SCs-EVs on axonal elongation was examined in vitro and in vivo. High throughput miRNA sequencing was performed to compare the differential miRNA profiles between MS-SCs-EVs and SCs-EVs. The functional role of differentially expressed miRNAs on neurite extension in MS-SCs-EVs was examined. Also, the putative target genes of differentially expressed miRNAs in MS-SCs-EVs were predicted by bioinformatics tools, and the regulatory effect of those miRNAs on putative target genes was validated both in vitro and in vivo. Results: The MS-SCs-EVs showed an average size of 137.52±1.77 nm, and could be internalized by dorsal root ganglion (DRG) neurons. Compared to SCs-EVs, MS-SCs-EVs showed a stronger ability to enhance neurite outgrowth in vitro and nerve regeneration in vivo. High throughput miRNA sequencing identified a number of differentially expressed miRNAs in MS-SCs-EVs. Further analysis of those EV-miRNAs demonstrated that miR-23b-3p played a predominant role in MS-SCs-EVs since its deprivation abolished their enhanced axonal elongation. Furthermore, we identified neuropilin 1 (Nrp1) in neurons as the target gene of miR-23b-3p in MS-SCs-EVs. This observation was supported by the evidence that miR-23b-3p could decrease Nrp1-3'-UTR-WT luciferase activity in vitro and down-regulate Nrp1 expression in neurons. Conclusion: Our findings suggested that mechanical stimuli are capable of modulating the intercellular communication between neurons and SCs by altering miRNA composition in MS-SCs-EVs. Transfer of miR-23b-3p by MS-SCs-EVs from mechanically stimulated SCs to neurons decreased neuronal Nrp1 expression, which was responsible, at least in part, for the beneficial effect of MS-SCs-EVs on axonal regeneration. Our results highlighted the potential therapeutic value of MS-SCs-EVs and miR-23b-3p-enriched EVs in peripheral nerve injury repair.
Collapse
Affiliation(s)
- Bing Xia
- Department of Orthopedics, Xijing Hospital, the Fourth Military Medical University, Xi'an, 710032, People's Republic of China
| | - Jianbo Gao
- Department of Orthopedics, Xijing Hospital, the Fourth Military Medical University, Xi'an, 710032, People's Republic of China
| | - Shengyou Li
- Department of Orthopedics, Xijing Hospital, the Fourth Military Medical University, Xi'an, 710032, People's Republic of China
| | - Liangliang Huang
- Department of Orthopedics, the General Hospital of Central Theater Command of People's Liberation Army, Wuhan, 430070, People's Republic of China
| | - Lei Zhu
- Department of Spine Surgery, Honghui Hospital Affiliated to Medical School of Xi'an Jiaotong University, Xi'an Shaanxi, 710054, People's Republic of China
| | - Teng Ma
- Department of Orthopedics, Xijing Hospital, the Fourth Military Medical University, Xi'an, 710032, People's Republic of China
| | - Laihe Zhao
- Department of Orthopedics, Xijing Hospital, the Fourth Military Medical University, Xi'an, 710032, People's Republic of China
| | - Yujie Yang
- Department of Orthopedics, Xijing Hospital, the Fourth Military Medical University, Xi'an, 710032, People's Republic of China
| | - Kai Luo
- Department of Orthopedics, the 985th Hospital People's Liberation Army Joint Logistics Support Force, Taiyuan, 030000, People's Republic of China
| | - Xiaowei Shi
- Department of Orthopedics, Xijing Hospital, the Fourth Military Medical University, Xi'an, 710032, People's Republic of China
| | - Liangwei Mei
- Department of Orthopedics, Xijing Hospital, the Fourth Military Medical University, Xi'an, 710032, People's Republic of China
| | - Hao Zhang
- Department of Spinal Surgery, the People's Hospital of Longhua District, Shenzhen, 518109, People's Republic of China
| | - Yi Zheng
- Department of Orthopedics, Xijing Hospital, the Fourth Military Medical University, Xi'an, 710032, People's Republic of China
| | - Lei Lu
- Department of Oral Anatomy and Physiology, State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of Stomatology, School of Stomatology, the Fourth Military Medical University, Xi'an, 710032, People's Republic of China
| | - Zhuojing Luo
- Department of Orthopedics, Xijing Hospital, the Fourth Military Medical University, Xi'an, 710032, People's Republic of China
| | - Jinghui Huang
- Department of Orthopedics, Xijing Hospital, the Fourth Military Medical University, Xi'an, 710032, People's Republic of China
| |
Collapse
|
14
|
Bai WF, Zhang Y, Xu W, Li W, Li M, Yuan F, Luo X, Zhang M. Isolation and Characterization of Neural Progenitor Cells From Bone Marrow in Cell Replacement Therapy of Brain Injury. Front Cell Neurosci 2020; 14:49. [PMID: 32226361 PMCID: PMC7080866 DOI: 10.3389/fncel.2020.00049] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Accepted: 02/21/2020] [Indexed: 12/27/2022] Open
Abstract
Many studies supported that bone marrow mesenchymal stem cells (BM-MSCs) can differentiate into neural cells, but few researchers detected mature and function of nerve cells, especially in vivo study. Some researchers even suggested that BM-MSCs transplantation would not be able to differentiate into functional neural cells. To figure out the dispute, this study examined bone marrow-derived sphere-like cells, harvested via neural stem cell suspension culture, then identified as bone marrow-derived neural progenitor cells (BM-NPCs) by finding the expression of neural progenitor cells genes and proteins, neural progenitor cells characteristic and nerve cell differentiation induced through both methods. Moreover, BM-NPCs transplantation showed long-term survival and improved the ethological and histological indexes of brain injury rats, demonstrating functional nervous cells differentiated from BM-NPCs. These in vitro and in vivo results confirmed BM-NPCs differentiating into mature and functional nerve cells. This study provided valuable experimental data for BM-NPCs, suggesting a potential alternative treatment of central nervous injury disease.
Collapse
Affiliation(s)
- Wen-fang Bai
- Department of Rehabilitation Medicine, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangdong Provincial Institute of Geriatrics, Guangzhou, China
| | - Yuling Zhang
- School of Medical Instrument and Food Engineering, The University of Shanghai for Science and Technology, Shanghai, China
- Stroke Biological Recovery Laboratory, Spaulding Rehabilitation Hospital, a Teaching Affiliate of Harvard Medical School, Charlestown, MA, United States
| | - Weicheng Xu
- Department of Rehabilitation Medicine, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangdong Provincial Institute of Geriatrics, Guangzhou, China
| | - Weikun Li
- Department of Rehabilitation Medicine, Zengcheng District People’s Hospital of Guangzhou, Guangzhou, China
| | - Meihui Li
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China
| | - Fengying Yuan
- Department of Rehabilitation Medicine, Guangdong Provincial Hospital of Integrated Traditional Chinese and Western Medicine, Foshan, China
| | - Xun Luo
- Kerry Rehabilitation Medicine Research Institute, Shenzhen, China
- Shenzhen Sanming Project Group, Spaulding Rehabilitation Hospital, a Teaching Affiliate of Harvard Medical School, Charlestown, MA, United States
| | - Mingsheng Zhang
- Department of Rehabilitation Medicine, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangdong Provincial Institute of Geriatrics, Guangzhou, China
- *Correspondence: Mingsheng Zhang
| |
Collapse
|
15
|
Electrical stimulation affects neural stem cell fate and function in vitro. Exp Neurol 2019; 319:112963. [PMID: 31125549 DOI: 10.1016/j.expneurol.2019.112963] [Citation(s) in RCA: 121] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Revised: 04/29/2019] [Accepted: 05/19/2019] [Indexed: 11/22/2022]
Abstract
Electrical stimulation (ES) has been applied in cell culture system to enhance neural stem cell (NSC) proliferation, neuronal differentiation, migration, and integration. According to the mechanism of its function, ES can be classified into induced electrical (EFs) and electromagnetic fields (EMFs). EFs guide axonal growth and induce directional cell migration, whereas EMFs promote neurogenesis and facilitates NSCs to differentiate into functional neurons. Conductive nanomaterials have been used as functional scaffolds to provide mechanical support and biophysical cues in guiding neural cell growth and differentiation and building complex neural tissue patterns. Nanomaterials may have a combined effect of topographical and electrical cues on NSC migration and differentiation. Electrical cues may promote NSC neurogenesis via specific ion channel activation, such as SCN1α and CACNA1C. To accelerate the future application of ES in preclinical research, we summarized the specific setting, such as current frequency, intensity, and stimulation duration used in various ES devices, as well as the nanomaterials involved, in this review with the possible mechanisms elucidated. This review can be used as a checklist for ES work in stem cell research to enhance the translational process of NSCs in clinical application.
Collapse
|
16
|
Kocaman A, Altun G, Kaplan AA, Deniz ÖG, Yurt KK, Kaplan S. Genotoxic and carcinogenic effects of non-ionizing electromagnetic fields. ENVIRONMENTAL RESEARCH 2018; 163:71-79. [PMID: 29427953 DOI: 10.1016/j.envres.2018.01.034] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Revised: 01/14/2018] [Accepted: 01/23/2018] [Indexed: 05/06/2023]
Abstract
New technologies in electronics and communications are continually emerging. An increasing use of these electronic devices such as mobile phone, computer, wireless fidelity connectors or cellular towers is raising questions concerning whether they have an adverse effect on the body. Exposure to electromagnetic fields (EMF) is frequently suggested to have adverse health effects on humans and other organisms. This idea has been reported in many studies. In contrast, the therapeutic effects of EMF on different organs have also been reported. Research findings are inconsistent. This has given rise to very profound discrepancies. The duration and frequency of mobile phone calls and the association observed with various health effects has raised serious concerns due to the frequency with which these devices are used and the way they are held close to the head. The present review assesses the results of in vitro, in vivo, experimental, and epidemiological studies. The purpose of the study is to assess data concerning the carcinogenic and genotoxic effects of non-ionizing EMF. The major genotoxic and carcinogenic effects of EMF, divided into subsections as low frequency effects and radiofrequency effects, were reviewed. The inconsistent results between similar studies and the same research groups have made it very difficult to make any comprehensive interpretation. However, evaluation of current studies suggests that EMF may represent a serious source of concern and may be hazardous to living organisms.
Collapse
Affiliation(s)
- Adem Kocaman
- Department of Histology and Embryology, Medical Faculty, Ondokuz Mayıs University, Samsun, Turkey.
| | - Gamze Altun
- Department of Histology and Embryology, Medical Faculty, Ondokuz Mayıs University, Samsun, Turkey
| | - Arife Ahsen Kaplan
- Department of Histology and Embryology, Medical Faculty, Ondokuz Mayıs University, Samsun, Turkey
| | - Ömür Gülsüm Deniz
- Department of Histology and Embryology, Medical Faculty, Ondokuz Mayıs University, Samsun, Turkey
| | - Kıymet Kübra Yurt
- Department of Histology and Embryology, Medical Faculty, Ondokuz Mayıs University, Samsun, Turkey
| | - Süleyman Kaplan
- Department of Histology and Embryology, Medical Faculty, Ondokuz Mayıs University, Samsun, Turkey
| |
Collapse
|
17
|
Zuo H, Liu X, Wang D, Li Y, Xu X, Peng R, Song T. RKIP-Mediated NF-κB Signaling is involved in ELF-MF-mediated improvement in AD rat. Int J Med Sci 2018; 15:1658-1666. [PMID: 30588189 PMCID: PMC6299414 DOI: 10.7150/ijms.28411] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Accepted: 10/12/2018] [Indexed: 12/24/2022] Open
Abstract
In a previous study, we reported the positive effects of extremely low frequency electromagnetic field (ELF-MF) exposure on Alzheimer's disease (AD) rats; however, the underlying mechanism remains unclear. In addition, we found that Raf-1 kinase inhibitor protein (RKIP) was downregulated by microwave exposure in the rat hippocampus. Our hypothesis was that RKIP-mediated NF-κB pathway signaling is involved in the effect of ELF-MF on the AD rat. In this study, D-galactose intraperitoneal (50 mg/kg/d for 42 d) and Aβ25-35 hippocampal (5 μL/unilateral, bilateral, single-dose) injection were implemented to establish an AD rat model. Animals were exposed to 50 Hz and 400 µT ELF-MF for 60 continuous days. The spatial memory ability of the rat was then tested using the Morris water maze. Protein expression and interaction were detected by western blotting and co-immunoprecipitation for RKIP-mediated NF-κB pathway factors. The results showed that ELF-MF exposure partially improved the cognitive disorder, upregulated the levels of RKIP, TAK1, and the RKIP/TAK1 interaction, but downregulated p-IKK levels in AD rats. These results indicated that RKIP-mediated NF-κB pathway signaling plays an important role in the ELF-MF exposure-mediated improvements in the AD rat. Our study suggested that ELF-MF exposure might have a potential therapeutic value for AD. Further in depth studies are required in the future.
Collapse
Affiliation(s)
- Hongyan Zuo
- Department of Experimental Pathology, Beijing Institute of Radiation Medicine, Beijing, China
| | - Xiao Liu
- Department of Experimental Pathology, Beijing Institute of Radiation Medicine, Beijing, China
| | - Dewen Wang
- Department of Experimental Pathology, Beijing Institute of Radiation Medicine, Beijing, China
| | - Yang Li
- Department of Experimental Pathology, Beijing Institute of Radiation Medicine, Beijing, China
| | - Xinping Xu
- Department of Experimental Pathology, Beijing Institute of Radiation Medicine, Beijing, China
| | - Ruiyun Peng
- Department of Experimental Pathology, Beijing Institute of Radiation Medicine, Beijing, China
| | - Tao Song
- Beijing Key Laboratory of Bioelectromagnetism, Institute of Electrical Engineering, Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
18
|
Seo N, Lee SH, Ju KW, Woo J, Kim B, Kim S, Jahng JW, Lee JH. Low-frequency pulsed electromagnetic field pretreated bone marrow-derived mesenchymal stem cells promote the regeneration of crush-injured rat mental nerve. Neural Regen Res 2018; 13:145-153. [PMID: 29451219 PMCID: PMC5840980 DOI: 10.4103/1673-5374.224383] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Bone marrow-derived mesenchymal stem cells (BMSCs) have been shown to promote the regeneration of injured peripheral nerves. Pulsed electromagnetic field (PEMF) reportedly promotes the proliferation and neuronal differentiation of BMSCs. Low-frequency PEMF can induce the neuronal differentiation of BMSCs in the absence of nerve growth factors. This study was designed to investigate the effects of low-frequency PEMF pretreatment on the proliferation and function of BMSCs and the effects of low-frequency PEMF pre-treated BMSCs on the regeneration of injured peripheral nerve using in vitro and in vivo experiments. In in vitro experiments, quantitative DNA analysis was performed to determine the proliferation of BMSCs, and reverse transcription-polymerase chain reaction was performed to detect S100 (Schwann cell marker), glial fibrillary acidic protein (astrocyte marker), and brain-derived neurotrophic factor and nerve growth factor (neurotrophic factors) mRNA expression. In the in vivo experiments, rat models of crush-injured mental nerve established using clamp method were randomly injected with low-frequency PEMF pretreated BMSCs, unpretreated BMSCs or PBS at the injury site (1 × 106 cells). DiI-labeled BMSCs injected at the injury site were counted under the fluorescence microscope to determine cell survival. One or two weeks after cell injection, functional recovery of the injured nerve was assessed using the sensory test with von Frey filaments. Two weeks after cell injection, axonal regeneration was evaluated using histomorphometric analysis and retrograde labeling of trigeminal ganglion neurons. In vitro experiment results revealed that low-frequency PEMF pretreated BMSCs proliferated faster and had greater mRNA expression of growth factors than unpretreated BMSCs. In vivo experiment results revealed that compared with injection of unpretreated BMSCs, injection of low-frequency PEMF pretreated BMSCs led to higher myelinated axon count and axon density and more DiI-labeled neurons in the trigeminal ganglia, contributing to rapider functional recovery of injured mental nerve. These findings suggest that low-frequency PEMF pretreatment is a promising approach to enhance the efficacy of cell therapy for peripheral nerve injury repair.
Collapse
Affiliation(s)
- NaRi Seo
- Department of Oral and Maxillofacial Surgery, Graduate School of Dentistry, Seoul National University; Dental Research Institute, Seoul National University, Seoul, South Korea
| | - Sung-Ho Lee
- Department of Oral and Maxillofacial Surgery, Seoul National University Dental Hospital; Dental Research Institute, Seoul National University, Seoul, South Korea
| | - Kyung Won Ju
- Department of Oral and Maxillofacial Surgery, Seoul National University Dental Hospital; Dental Research Institute, Seoul National University, Seoul, South Korea
| | - JaeMan Woo
- Department of Oral and Maxillofacial Surgery, Seoul National University Dental Hospital, Seoul, South Korea
| | - BongJu Kim
- Clinical Translational Research Center for Dental Science (CTRC), Seoul National University Dental Hospital, Seoul, South Korea
| | - SoungMin Kim
- Department of Oral and Maxillofacial Surgery, Graduate School of Dentistry, Seoul National University; Department of Oral and Maxillofacial Surgery, Seoul National University Dental Hospital, Seoul, South Korea
| | - Jeong Won Jahng
- Dental Research Institute, Seoul National University, Seoul, South Korea
| | - Jong-Ho Lee
- Department of Oral and Maxillofacial Surgery, Graduate School of Dentistry, Seoul National University; Department of Oral and Maxillofacial Surgery, Seoul National University Dental Hospital; Dental Research Institute, Seoul National University; Clinical Translational Research Center for Dental Science (CTRC), Seoul National University Dental Hospital, Seoul, South Korea
| |
Collapse
|
19
|
Liu Z, Zhu S, Liu L, Ge J, Huang L, Sun Z, Zeng W, Huang J, Luo Z. A magnetically responsive nanocomposite scaffold combined with Schwann cells promotes sciatic nerve regeneration upon exposure to magnetic field. Int J Nanomedicine 2017; 12:7815-7832. [PMID: 29123395 PMCID: PMC5661463 DOI: 10.2147/ijn.s144715] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Peripheral nerve repair is still challenging for surgeons. Autologous nerve transplantation is the acknowledged therapy; however, its application is limited by the scarcity of available donor nerves, donor area morbidity, and neuroma formation. Biomaterials for engineering artificial nerves, particularly materials combined with supportive cells, display remarkable promising prospects. Schwann cells (SCs) are the absorbing seeding cells in peripheral nerve engineering repair; however, the attenuated biologic activity restricts their application. In this study, a magnetic nanocomposite scaffold fabricated from magnetic nanoparticles and a biodegradable chitosan-glycerophosphate polymer was made. Its structure was evaluated and characterized. The combined effects of magnetic scaffold (MG) with an applied magnetic field (MF) on the viability of SCs and peripheral nerve injury repair were investigated. The magnetic nanocomposite scaffold showed tunable magnetization and degradation rate. The MGs synergized with the applied MF to enhance the viability of SCs after transplantation. Furthermore, nerve regeneration and functional recovery were promoted by the synergism of SCs-loaded MGs and MF. Based on the current findings, the combined application of MGs and SCs with applied MF is a promising therapy for the engineering of peripheral nerve regeneration.
Collapse
Affiliation(s)
- Zhongyang Liu
- Department of Orthopedics, Xijing Hospital, Fourth Military Medical University, Xi’an, Shaanxi
| | - Shu Zhu
- Department of Orthopedics, Xijing Hospital, Fourth Military Medical University, Xi’an, Shaanxi
| | - Liang Liu
- Department of Orthopedics, No 161 Hospital of PLA, Wuhan, Hubei
| | - Jun Ge
- Department of Orthopedics, No 323 Hospital of PLA, Xi’an, Shaanxi
- Department of Anatomy, Fourth Military Medical University, Xi’an, Shaanxi
| | - Liangliang Huang
- Department of Orthopedics, Xijing Hospital, Fourth Military Medical University, Xi’an, Shaanxi
| | - Zhen Sun
- Department of Orthopedics, Xijing Hospital, Fourth Military Medical University, Xi’an, Shaanxi
| | - Wen Zeng
- Department of Neurosurgery, Tangdu Hospital, Fourth Military Medical University, Xi’an, Shaanxi, People’s Republic of China
| | - Jinghui Huang
- Department of Orthopedics, Xijing Hospital, Fourth Military Medical University, Xi’an, Shaanxi
| | - Zhuojing Luo
- Department of Orthopedics, Xijing Hospital, Fourth Military Medical University, Xi’an, Shaanxi
| |
Collapse
|
20
|
Haghighat N, Abdolmaleki P, Parnian J, Behmanesh M. The expression of pluripotency and neuronal differentiation markers under the influence of electromagnetic field and nitric oxide. Mol Cell Neurosci 2017; 85:19-28. [PMID: 28843440 DOI: 10.1016/j.mcn.2017.08.005] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Revised: 08/05/2017] [Accepted: 08/21/2017] [Indexed: 02/06/2023] Open
Abstract
Nitric oxide (NO) is a diatomic free radical compound that as a secondary messenger contributes to cell physiological functions and its variations influence proteins activity and triggering intracellular signaling cascades. Low frequency electromagnetic field (EMF) alters the cell biology such as cell differentiation by targeting the plasma membrane and entering force to the ions and small electrical ligands. The effect of these chemical (NO) and physical (EMF) factors on the expression of the stemness and neuronal differentiation markers in rat bone marrow mesenchymal stem cells (BMSC) was investigated. The cells were treated with low (50micromolar) and high (1mM) concentrations of Deta-NO as a NO donor molecule and 50Hz low frequency EMF. The expression of pluripotency and neuronal differentiation genes and proteins was investigated using real time qPCR and Immunocytochemistry techniques. The simultaneous treatment of EMF with NO (1mM) led to the down-regulation of stemness markers expression and up-regulation of neuronal differentiation markers expression. Cell proliferation decreased and cell morphology changed which caused the majority of cells obtains neuronal protein markers in their cytoplasm. The decrease in the expression of neuronal differentiation Nestin and DCX markers without any change in the expression of pluripotency Oct4 marker (treated with low concentration of NO) indicates protection of stemness state in these cells. Treatment with NO demonstrated a double behavior. NO low concentration helped the cells protect the stemness state but NO high concentration plus EMF pushed cells into differentiation pathway.
Collapse
Affiliation(s)
- Nazanin Haghighat
- Department of Biophysics, Faculty of Biological Science, Tarbiat Modares University (TMU), POB 14115-154, Tehran, Iran
| | - Parviz Abdolmaleki
- Department of Biophysics, Faculty of Biological Science, Tarbiat Modares University (TMU), POB 14115-154, Tehran, Iran.
| | - Javad Parnian
- Department of Biotechnology, Iranian Research Organization for Science and Technology, Tehran, Iran
| | - Mehrdad Behmanesh
- Department of Genetics, Faculty of Biological Science, Tarbiat Modares University (TMU), POB 14115-154, Tehran, Iran
| |
Collapse
|
21
|
Kuzniar A, Laffeber C, Eppink B, Bezstarosti K, Dekkers D, Woelders H, Zwamborn APM, Demmers J, Lebbink JHG, Kanaar R. Semi-quantitative proteomics of mammalian cells upon short-term exposure to non-ionizing electromagnetic fields. PLoS One 2017; 12:e0170762. [PMID: 28234898 PMCID: PMC5325209 DOI: 10.1371/journal.pone.0170762] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Accepted: 01/10/2017] [Indexed: 01/26/2023] Open
Abstract
The potential effects of non-ionizing electromagnetic fields (EMFs), such as those emitted by power-lines (in extremely low frequency range), mobile cellular systems and wireless networking devices (in radio frequency range) on human health have been intensively researched and debated. However, how exposure to these EMFs may lead to biological changes underlying possible health effects is still unclear. To reveal EMF-induced molecular changes, unbiased experiments (without a priori focusing on specific biological processes) with sensitive readouts are required. We present the first proteome-wide semi-quantitative mass spectrometry analysis of human fibroblasts, osteosarcomas and mouse embryonic stem cells exposed to three types of non-ionizing EMFs (ELF 50 Hz, UMTS 2.1 GHz and WiFi 5.8 GHz). We performed controlled in vitro EMF exposures of metabolically labeled mammalian cells followed by reliable statistical analyses of differential protein- and pathway-level regulations using an array of established bioinformatics methods. Our results indicate that less than 1% of the quantitated human or mouse proteome responds to the EMFs by small changes in protein abundance. Further network-based analysis of the differentially regulated proteins did not detect significantly perturbed cellular processes or pathways in human and mouse cells in response to ELF, UMTS or WiFi exposure. In conclusion, our extensive bioinformatics analyses of semi-quantitative mass spectrometry data do not support the notion that the short-time exposures to non-ionizing EMFs have a consistent biologically significant bearing on mammalian cells in culture.
Collapse
Affiliation(s)
- Arnold Kuzniar
- Department of Molecular Genetics, Cancer Genomics Netherlands, Erasmus University Medical Center, Rotterdam, The Netherlands
- Netherlands eScience Center, Amsterdam, The Netherlands
- * E-mail: (RK); (AK)
| | - Charlie Laffeber
- Department of Molecular Genetics, Cancer Genomics Netherlands, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Berina Eppink
- Department of Molecular Genetics, Cancer Genomics Netherlands, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Karel Bezstarosti
- Proteomics Center, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Dick Dekkers
- Proteomics Center, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Henri Woelders
- Wageningen Livestock Research, Wageningen, The Netherlands
| | | | - Jeroen Demmers
- Proteomics Center, Erasmus University Medical Center, Rotterdam, The Netherlands
- Netherlands Proteomics Center, Rotterdam, The Netherlands
| | - Joyce H. G. Lebbink
- Department of Molecular Genetics, Cancer Genomics Netherlands, Erasmus University Medical Center, Rotterdam, The Netherlands
- Department of Radiation Oncology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Roland Kanaar
- Department of Molecular Genetics, Cancer Genomics Netherlands, Erasmus University Medical Center, Rotterdam, The Netherlands
- Department of Radiation Oncology, Erasmus University Medical Center, Rotterdam, The Netherlands
- * E-mail: (RK); (AK)
| |
Collapse
|
22
|
Bai WF, Xu WC, Zhu HX, Huang H, Wu B, Zhang MS. Efficacy of 50 Hz electromagnetic fields on human epidermal stem cell transplantation seeded in collagen sponge scaffolds for wound healing in a murine model. Bioelectromagnetics 2017; 38:204-212. [PMID: 28106913 DOI: 10.1002/bem.22029] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2015] [Accepted: 12/03/2016] [Indexed: 11/11/2022]
Abstract
To explore the possible efficacy of electromagnetic fields (EMF) for skin tissue engineering, effects of EMF exposure on epidermal stem cells (ESC) seeded in collagen sponge scaffolds for wound healing in a murine model were investigated. The wound models of a full-thickness defect established with 36 7 ∼ 8-week-old nude mice were randomly divided into three groups: a control group, an ESC-only group, and an ESC with EMF exposure group (frequency of 50 Hz, magnetic induction of 5 mT, 60 min per day for 20 days). ESC were separated from human foreskin and cultured in vitro, and then transplanted with collagen sponge scaffolds as a delivery vehicle to wounds of the ESC-only group, and ESC with EMF exposure group was exposed to EMF after ESC transplantation. Effects of EMF on morphological changes and expression of β1 integrin in regenerated skins were observed. Wound healing rates and healing times were collected to evaluate the efficacy of repairment. Results showed that human ESC were successfully transplanted to nude mice, which facilitated the formation of intact skin on nude mice. In contrast to other groups, the wound healing of ESC with EMF exposure group was the fastest (P < 0.05), the structure of regenerated skins was more mature, and it contained more continuity in the number of viable cell layers and rich hair follicles' structure. These results suggest that the use of 50 Hz EMF as a non-invasive treatment can accelerate wound healing of ESC transplantation, and restore structural integrity of regenerated skin. Bioelectromagnetics. 38:204-212,2017. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Wen-Fang Bai
- Jinan University, Guangzhou, China.,Department of Rehabilitation Medicine, Institute of Geriatric Medicine, Guangdong Academy of Medical Sciences, Guangdong General Hospital, Guangzhou, China
| | - Wei-Cheng Xu
- Department of Rehabilitation Medicine, Institute of Geriatric Medicine, Guangdong Academy of Medical Sciences, Guangdong General Hospital, Guangzhou, China
| | - Hong-Xiang Zhu
- Department of Rehabilitation Medicine, Institute of Geriatric Medicine, Guangdong Academy of Medical Sciences, Guangdong General Hospital, Guangzhou, China
| | - Hong Huang
- School of Information, University of South Florida, Tampa, Florida
| | - Bo Wu
- Department of Rehabilitation Medicine, Institute of Geriatric Medicine, Guangdong Academy of Medical Sciences, Guangdong General Hospital, Guangzhou, China
| | - Ming-Sheng Zhang
- Department of Rehabilitation Medicine, Institute of Geriatric Medicine, Guangdong Academy of Medical Sciences, Guangdong General Hospital, Guangzhou, China
| |
Collapse
|
23
|
Choi YK, Urnukhsaikhan E, Yoon HH, Seo YK, Cho H, Jeong JS, Kim SC, Park JK. Combined effect of pulsed electromagnetic field and sound wave on In vitro and In vivo neural differentiation of human mesenchymal stem cells. Biotechnol Prog 2016; 33:201-211. [PMID: 27790871 DOI: 10.1002/btpr.2389] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Revised: 08/10/2016] [Indexed: 12/21/2022]
Abstract
Biophysical wave stimulus has been used as an effective tool to promote cellular maturation and differentiation in the construction of engineered tissue. Pulsed electromagnetic fields (PEMFs) and sound waves have been selected as effective stimuli that can promote neural differentiation. The aim of this study was to investigate the synergistic effect of PEMFs and sound waves on the neural differentiation potential in vitro and in vivo using human bone marrow mesenchymal stem cells (hBM-MSCs). In vitro, neural-related genes in hBM-MSCs were accelerated by the combined exposure to both waves more than by individual exposure to PEMFs or sound waves. The combined wave also up-regulated the expression of neural and synaptic-related proteins in a three-dimensional (3-D) culture system through the phosphorylation of extracellular signal-related kinase. In a mouse model of photochemically induced ischemia, exposure to the combined wave reduced the infarction volume and improved post-injury behavioral activity. These results indicate that a combined stimulus of biophysical waves, PEMFs and sound can enhance and possibly affect the differentiation of MSCs into neural cells. Our study is meaningful for highlighting the potential of combined wave for neurogenic effects and providing new therapeutic approaches for neural cell therapy. © 2016 American Institute of Chemical Engineers Biotechnol. Prog., 33:201-211, 2017.
Collapse
Affiliation(s)
- Yun-Kyong Choi
- Dept. of Medical Biotechnology, Dongguk University, Seoul, Korea
| | | | - Hee-Hoon Yoon
- Dongguk University Research Inst. of Biotechnology, Seoul, Korea
| | - Young-Kwon Seo
- Dept. of Medical Biotechnology, Dongguk University, Seoul, Korea
| | - Hyunjin Cho
- Dongguk University Research Inst. of Biotechnology, Seoul, Korea
| | - Jong-Seob Jeong
- Dept. of Medical Biotechnology, Dongguk University, Seoul, Korea
| | - Soo-Chan Kim
- Graduate School of Bio and Information Technology, Hankyong National University, Anseong-si, Kyonggi-do, Korea
| | - Jung-Keug Park
- Dept. of Medical Biotechnology, Dongguk University, Seoul, Korea
| |
Collapse
|
24
|
Kavand H, Haghighipour N, Zeynali B, Seyedjafari E, Abdemami B. Extremely Low Frequency Electromagnetic Field in Mesenchymal Stem Cells Gene Regulation: Chondrogenic Markers Evaluation. Artif Organs 2016; 40:929-937. [PMID: 27086585 DOI: 10.1111/aor.12696] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2015] [Revised: 11/14/2015] [Accepted: 12/23/2015] [Indexed: 01/02/2023]
Abstract
There is little evidence demonstrating the effects of electromagnetic fields (EMFs) generated within the biological entity and the effect of extrinsic fields on cellular programing. Taking the path of the more studied stimuli into attention, mechanical forces, it could be understood that nonchemical factors play a consequential role in transcriptional regulatory networks. Cartilaginous tissue consists of collagen protein that is considered as a piezoelectric substrate and is influenced by electric fields making chondrogenic specific genes an exciting candidate for bioelectromagnetic studies. As electromagnetic properties highly depend on the frequencies applied, this study delves into the ability of two EMFs with the frequency of 25 Hz and 50 Hz in inducing SOX9 and COL2 gene expressions in a three-dimensional (3D) mesenchymal stem cell (MSC)-alginate construct. Cell-alginate beads were divided into six groups and treated for a time period of 21 days. To determine the results, qualitative and quantitative data were both reviewed. On observation of real-time polymerase chain reaction (PCR) data, it was apparent that TGF-β1 treatment had a greater COL2 and SOX9 gene expression impact on MSCs compared to pulsed electromagnetic field (PEMF) treatments alone. COL2 was shown to have a greater transcriptional tendency to PEMF, whereas under defined electromagnetic parameters applied in this study, no significant difference was detected in SOX9 gene expressions compared to the control group. PEMF co-treatments enhanced the deposition of extracellular matrix molecules, as the matrix-rich beads were positively stained by Alcian blue. This genre of study is the venue for the control and healing of connective tissue defects.
Collapse
Affiliation(s)
- Hanie Kavand
- National Cell Bank of Iran, Pasteur Institute of Iran, Tehran, Iran
- Department of Cell and Molecular Biology, University of Tehran, Tehran, Iran
| | | | - Bahman Zeynali
- Developmental Biology Lab, School of Biology, University of Tehran, Tehran, Iran
| | - Ehsan Seyedjafari
- Department of Biotechnology, College of Science, University of Tehran, Tehran, Iran
| | - Baharak Abdemami
- National Cell Bank of Iran, Pasteur Institute of Iran, Tehran, Iran
| |
Collapse
|
25
|
Ross CL, Syed I, Smith TL, Harrison BS. The regenerative effects of electromagnetic field on spinal cord injury. Electromagn Biol Med 2016; 36:74-87. [DOI: 10.3109/15368378.2016.1160408] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
|
26
|
Iachininoto MG, Camisa V, Leone L, Pinto R, Lopresto V, Merla C, Giorda E, Carsetti R, Zaffina S, Podda MV, Teofili L, Grassi C. Effects of exposure to gradient magnetic fields emitted by nuclear magnetic resonance devices on clonogenic potential and proliferation of human hematopoietic stem cells. Bioelectromagnetics 2016; 37:201-11. [PMID: 26992028 DOI: 10.1002/bem.21967] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2015] [Accepted: 02/26/2016] [Indexed: 01/29/2023]
Abstract
This study investigates effects of gradient magnetic fields (GMFs) emitted by magnetic resonance imaging (MRI) devices on hematopoietic stem cells. Field measurements were performed to assess exposure to GMFs of staff working at 1.5 T and 3 T MRI units. Then an exposure system reproducing measured signals was realized to expose in vitro CD34+ cells to GMFs (1.5 T-protocol and 3 T-protocol). CD34+ cells were obtained by Fluorescence Activated Cell Sorting from six blood donors and three MRI-exposed workers. Blood donor CD34+ cells were exposed in vitro for 72 h to 1.5 T or 3 T-protocol and to sham procedure. Cells were then cultured and evaluated in colony forming unit (CFU)-assay up to 4 weeks after exposure. Results showed that in vitro GMF exposure did not affect cell proliferation but instead induced expansion of erythroid and monocytes progenitors soon after exposure and for the subsequent 3 weeks. No decrease of other clonogenic cell output (i.e., CFU-granulocyte/erythroid/macrophage/megakaryocyte and CFU-granulocyte/macrophage) was noticed, nor exposed CD34+ cells underwent the premature exhaustion of their clonogenic potential compared to sham-exposed controls. On the other hand, pilot experiments showed that CD34+ cells exposed in vivo to GMFs (i.e., samples from MRI workers) behaved in culture similarly to sham-exposed CD34+ cells, suggesting that other cells and/or microenvironment factors might prevent GMF effects on hematopoietic stem cells in vivo. Accordingly, GMFs did not affect the clonogenic potential of umbilical cord blood CD34+ cells exposed in vitro together with the whole mononuclear cell fraction.
Collapse
Affiliation(s)
| | - Vincenzo Camisa
- Occupational Medicine/Health Technology Assessment and Safety Research Unit, Clinical-Technological Innovations Research Area-Bambino Gesù Children's Hospital IRCCS, Rome, Italy
| | - Lucia Leone
- Institute of Human Physiology, Università Cattolica, Rome, Italy
| | - Rosanna Pinto
- ENEA, Italian Agency for New Technologies, Energy, and Sustainable Economic Development, Division of Health Protection Technologies, Casaccia Research Centre, Rome, Italy
| | - Vanni Lopresto
- ENEA, Italian Agency for New Technologies, Energy, and Sustainable Economic Development, Division of Health Protection Technologies, Casaccia Research Centre, Rome, Italy
| | - Caterina Merla
- ENEA, Italian Agency for New Technologies, Energy, and Sustainable Economic Development, Division of Health Protection Technologies, Casaccia Research Centre, Rome, Italy
| | - Ezio Giorda
- Immunology Unit, Immunology and Pharmacotherapy Research Area-Bambino Gesù Children's Hospital IRCCS, Rome, Italy
| | - Rita Carsetti
- Immunology Unit, Immunology and Pharmacotherapy Research Area-Bambino Gesù Children's Hospital IRCCS, Rome, Italy
| | - Salvatore Zaffina
- Occupational Medicine/Health Technology Assessment and Safety Research Unit, Clinical-Technological Innovations Research Area-Bambino Gesù Children's Hospital IRCCS, Rome, Italy
| | | | - Luciana Teofili
- Department of Transfusion Medicine, Institute of Hematology, Università Cattolica, Rome, Italy
| | - Claudio Grassi
- Institute of Human Physiology, Università Cattolica, Rome, Italy.,San Raffaele Pisana Scientific Institute for Research, Hospitalization and Health Care, Rome, Italy
| |
Collapse
|
27
|
Urnukhsaikhan E, Cho H, Mishig-Ochir T, Seo YK, Park JK. Pulsed electromagnetic fields promote survival and neuronal differentiation of human BM-MSCs. Life Sci 2016; 151:130-138. [PMID: 26898125 DOI: 10.1016/j.lfs.2016.02.066] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2015] [Revised: 02/15/2016] [Accepted: 02/16/2016] [Indexed: 12/11/2022]
Abstract
Pulsed electromagnetic fields (PEMF) are known to affect biological properties such as differentiation, regulation of transcription factor and cell proliferation. However, the cell-protective effect of PEMF exposure is largely unknown. The aim of this study is to understand the mechanisms underlying PEMF-mediated suppression of apoptosis and promotion of survival, including PEMF-induced neuronal differentiation. Treatment of induced human BM-MSCs with PEMF increased the expression of neural markers such as NF-L, NeuroD1 and Tau. Moreover, treatment of induced human BM-MSCs with PEMF greatly decreased cell death in a dose- and time-dependent manner. There is evidence that Akt and Ras are involved in neuronal survival and protection. Activation of Akt and Ras results in the regulation of survival proteins such as Bad and Bcl-xL. Thus, the Akt/Ras signaling pathway may be a desirable target for enhancing cell survival and treatment of neurological disease. Our analyses indicated that PEMF exposure dramatically increased the activity of Akt, Rsk, Creb, Erk, Bcl-xL and Bad via phosphorylation. PEMF-dependent cell protection was reversed by pretreatment with LY294002, a specific inhibitor of phosphatidylinositol 3-kinase (PI3K). Our data suggest that the PI3K/Akt/Bad signaling pathway may be a possible mechanism for the cell-protective effects of PEMF.
Collapse
Affiliation(s)
| | - Hyunjin Cho
- Dongguk University Research Institute of Biotechnology, Republic of Korea
| | | | - Young-Kwon Seo
- Department of Medical Biotechnology, Dongguk University, Seoul, Republic of Korea
| | - Jung-Kueg Park
- Department of Medical Biotechnology, Dongguk University, Seoul, Republic of Korea
| |
Collapse
|
28
|
Yang G, Ren Z, Mei YA. Exposure to 50 Hz magnetic field modulates GABAA currents in cerebellar granule neurons through an EP receptor-mediated PKC pathway. J Cell Mol Med 2015; 19:2413-22. [PMID: 26176998 PMCID: PMC4594682 DOI: 10.1111/jcmm.12626] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2015] [Accepted: 05/12/2015] [Indexed: 12/31/2022] Open
Abstract
Previous work from both our lab and others have indicated that exposure to 50 Hz magnetic fields (ELF-MF) was able to modify ion channel functions. However, very few studies have investigated the effects of MF on γ-aminobutyric acid (GABA) type A receptors (GABA(A) Rs) channel functioning, which are fundamental to overall neuronal excitability. Here, our major goal is to reveal the potential effects of ELF-MF on GABA(A) Rs activity in rat cerebellar granule neurons (CGNs). Our results indicated that exposing CGNs to 1 mT ELF-MF for 60 min. significantly increased GABA(A) R currents without modifying sensitivity to GABA. However, activation of PKA by db-cAMP failed to do so, but led to a slight decrease instead. On the other hand, PKC activation or inhibition by PMA or Bis and Docosahexaenoic acid (DHA) mimicked or eliminated the field-induced-increase of GABA(A) R currents. Western blot analysis indicated that the intracellular levels of phosphorylated PKC (pPKC) were significantly elevated after 60 min. of ELF-MF exposure, which was subsequently blocked by application of DHA or EP1 receptor-specific (prostaglandin E receptor 1) antagonist (SC19220), but not by EP2-EP4 receptor-specific antagonists. SC19220 also significantly inhibited the ELF-MF-induced elevation on GABA(A) R currents. Together, these data obviously demonstrated for the first time that neuronal GABA(A) currents are significantly increased by ELF-MF exposure, and also suggest that these effects are mediated via an EP1 receptor-mediated PKC pathway. Future work will focus on a more comprehensive analysis of the physiological and/or pathological consequences of these effects.
Collapse
Affiliation(s)
- Guang Yang
- School of Life Sciences, Institute of Brain Science and State Key Laboratory of Medical Neurobiology, Fudan UniversityShanghai, China
| | - Zhen Ren
- School of Life Sciences, Institute of Brain Science and State Key Laboratory of Medical Neurobiology, Fudan UniversityShanghai, China
| | - Yan-Ai Mei
- School of Life Sciences, Institute of Brain Science and State Key Laboratory of Medical Neurobiology, Fudan UniversityShanghai, China
| |
Collapse
|
29
|
Leone L, Podda MV, Grassi C. Impact of electromagnetic fields on stem cells: common mechanisms at the crossroad between adult neurogenesis and osteogenesis. Front Cell Neurosci 2015; 9:228. [PMID: 26124705 PMCID: PMC4466452 DOI: 10.3389/fncel.2015.00228] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Accepted: 05/31/2015] [Indexed: 12/18/2022] Open
Abstract
In the recent years adult neural and mesenchymal stem cells have been intensively investigated as effective resources for repair therapies. In vivo and in vitro studies have provided insights on the molecular mechanisms underlying the neurogenic and osteogenic processes in adulthood. This knowledge appears fundamental for the development of targeted strategies to manipulate stem cells. Here we review recent literature dealing with the effects of electromagnetic fields on stem cell biology that lends support to their use as a promising tool to positively influence the different steps of neurogenic and osteogenic processes. We will focus on recent studies revealing that extremely-low frequency electromagnetic fields enhance adult hippocampal neurogenesis by inducing epigenetic modifications on the regulatory sequences of genes responsible for neural stem cell proliferation and neuronal differentiation. In light of the emerging critical role played by chromatin modifications in maintaining the stemness as well as in regulating stem cell differentiation, we will also attempt to exploit epigenetic changes that can represent common targets for electromagnetic field effects on neurogenic and osteogenic processes.
Collapse
Affiliation(s)
- Lucia Leone
- Institute of Human Physiology, Medical School, Università Cattolica del Sacro Cuore Rome, Italy
| | - Maria Vittoria Podda
- Institute of Human Physiology, Medical School, Università Cattolica del Sacro Cuore Rome, Italy
| | - Claudio Grassi
- Institute of Human Physiology, Medical School, Università Cattolica del Sacro Cuore Rome, Italy
| |
Collapse
|
30
|
Liu X, Zuo H, Wang D, Peng R, Song T, Wang S, Xu X, Gao Y, Li Y, Wang S, Wang L, Zhao L. Improvement of spatial memory disorder and hippocampal damage by exposure to electromagnetic fields in an Alzheimer's disease rat model. PLoS One 2015; 10:e0126963. [PMID: 25978363 PMCID: PMC4433192 DOI: 10.1371/journal.pone.0126963] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2014] [Accepted: 04/09/2015] [Indexed: 01/05/2023] Open
Abstract
Although some epidemiological investigations showed a potential association between long-term exposure of extremely low frequency electromagnetic fields (ELF-EMF) and Alzheimer’s disease (AD), no reasonable mechanism can explain this association, and the related animal experiments are rare. In this study, ELF-EMF exposure (50Hz 400µT 60d) combined with D-galactose intraperitoneal (50mg/kg, q.d., 42d) and Aβ25–35 hippocampal (5μl/unilateral, bilateral, single-dose) injection was implemented to establish a complex rat model. Then the effects of ELF-EMF exposure on AD development was studied by using the Morris water maze, pathological analysis, and comparative proteomics. The results showed that ELF-EMF exposure delayed the weight gain of rats, and partially improved cognitive and clinicopathologic symptoms of AD rats. The differential proteomic analysis results suggest that synaptic transmission, oxidative stress, protein degradation, energy metabolism, Tau aggregation, and inflammation involved in the effects mentioned above. Therefore, our findings indicate that certain conditions of ELF-EMF exposure could delay the development of AD in rats.
Collapse
Affiliation(s)
- Xiao Liu
- Department of Experimental Pathology, Beijing Institute of Radiation Medicine, 27 Taiping Road, Haidian District, Beijing, China
| | - Hongyan Zuo
- Department of Experimental Pathology, Beijing Institute of Radiation Medicine, 27 Taiping Road, Haidian District, Beijing, China
- * E-mail: (HZ); (DW)
| | - Dewen Wang
- Department of Experimental Pathology, Beijing Institute of Radiation Medicine, 27 Taiping Road, Haidian District, Beijing, China
- * E-mail: (HZ); (DW)
| | - Ruiyun Peng
- Department of Experimental Pathology, Beijing Institute of Radiation Medicine, 27 Taiping Road, Haidian District, Beijing, China
| | - Tao Song
- Beijing Key Laboratory of Bioelectromagnetism, Institute of Electrical Engineering, Chinese Academy of Sciences, 6 North Second Street, Zhongguancun, Beijing, China
| | - Shuiming Wang
- Department of Experimental Pathology, Beijing Institute of Radiation Medicine, 27 Taiping Road, Haidian District, Beijing, China
| | - Xinping Xu
- Department of Experimental Pathology, Beijing Institute of Radiation Medicine, 27 Taiping Road, Haidian District, Beijing, China
| | - Yabing Gao
- Department of Experimental Pathology, Beijing Institute of Radiation Medicine, 27 Taiping Road, Haidian District, Beijing, China
| | - Yang Li
- Department of Experimental Pathology, Beijing Institute of Radiation Medicine, 27 Taiping Road, Haidian District, Beijing, China
| | - Shaoxia Wang
- Department of Experimental Pathology, Beijing Institute of Radiation Medicine, 27 Taiping Road, Haidian District, Beijing, China
| | - Lifeng Wang
- Department of Experimental Pathology, Beijing Institute of Radiation Medicine, 27 Taiping Road, Haidian District, Beijing, China
| | - Li Zhao
- Department of Experimental Pathology, Beijing Institute of Radiation Medicine, 27 Taiping Road, Haidian District, Beijing, China
| |
Collapse
|
31
|
Liu Z, Huang L, Liu L, Luo B, Liang M, Sun Z, Zhu S, Quan X, Yang Y, Ma T, Huang J, Luo Z. Activation of Schwann cells in vitro by magnetic nanocomposites via applied magnetic field. Int J Nanomedicine 2014; 10:43-61. [PMID: 25565803 PMCID: PMC4275057 DOI: 10.2147/ijn.s74332] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Schwann cells (SCs) are attractive seed cells in neural tissue engineering, but their application is limited by attenuated biological activities and impaired functions with aging. Therefore, it is important to explore an approach to enhance the viability and biological properties of SCs. In the present study, a magnetic composite made of magnetically responsive magnetic nanoparticles (MNPs) and a biodegradable chitosan–glycerophosphate polymer were prepared and characterized. It was further explored whether such magnetic nanocomposites via applied magnetic fields would regulate SC biological activities. The magnetization of the magnetic nanocomposite was measured by a vibrating sample magnetometer. The compositional characterization of the magnetic nanocomposite was examined by Fourier-transform infrared and X-ray diffraction. The tolerance of SCs to the magnetic fields was tested by flow-cytometry assay. The proliferation of cells was examined by a 5-ethynyl-2-deoxyuridine-labeling assay, a PrestoBlue assay, and a Live/Dead assay. Messenger ribonucleic acid of BDNF, GDNF, NT-3, and VEGF in SCs was assayed by quantitative real-time polymerase chain reaction. The amount of BDNF, GDNF, NT-3, and VEGF secreted from SCs was determined by enzyme-linked immunosorbent assay. It was found that magnetic nanocomposites containing 10% MNPs showed a cross-section diameter of 32.33±1.81 µm, porosity of 80.41%±0.72%, and magnetization of 5.691 emu/g at 8 kOe. The 10% MNP magnetic nanocomposites were able to support cell adhesion and spreading and further promote proliferation of SCs under magnetic field exposure. Interestingly, a magnetic field applied through the 10% MNP magnetic scaffold significantly increased the gene expression and protein secretion of BDNF, GDNF, NT-3, and VEGF. This work is the first stage in our understanding of how to precisely regulate the viability and biological properties of SCs in tissue-engineering grafts, which combined with additional molecular factors may lead to the development of new nerve grafts.
Collapse
Affiliation(s)
- Zhongyang Liu
- Department of Orthopaedics, Xijing Hospital, Fourth Military Medical University, Xi'an, People's Republic of China
| | - Liangliang Huang
- Department of Orthopaedics, Xijing Hospital, Fourth Military Medical University, Xi'an, People's Republic of China
| | - Liang Liu
- Department of Orthopaedics, Xijing Hospital, Fourth Military Medical University, Xi'an, People's Republic of China
| | - Beier Luo
- Department of Orthopaedics, Changhai Hospital, Second Military Medical University, Shanghai, People's Republic of China
| | - Miaomiao Liang
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, People's Republic of China
| | - Zhen Sun
- Department of Orthopaedics, Xijing Hospital, Fourth Military Medical University, Xi'an, People's Republic of China
| | - Shu Zhu
- Department of Orthopaedics, Xijing Hospital, Fourth Military Medical University, Xi'an, People's Republic of China
| | - Xin Quan
- Department of Orthopaedics, Xijing Hospital, Fourth Military Medical University, Xi'an, People's Republic of China
| | - Yafeng Yang
- Department of Orthopaedics, Xijing Hospital, Fourth Military Medical University, Xi'an, People's Republic of China
| | - Teng Ma
- Department of Orthopaedics, Xijing Hospital, Fourth Military Medical University, Xi'an, People's Republic of China
| | - Jinghui Huang
- Department of Orthopaedics, Xijing Hospital, Fourth Military Medical University, Xi'an, People's Republic of China
| | - Zhuojing Luo
- Department of Orthopaedics, Xijing Hospital, Fourth Military Medical University, Xi'an, People's Republic of China
| |
Collapse
|
32
|
Wang Q, Wu W, Han X, Zheng A, Lei S, Wu J, Chen H, He C, Luo F, Liu X. Osteogenic differentiation of amniotic epithelial cells: synergism of pulsed electromagnetic field and biochemical stimuli. BMC Musculoskelet Disord 2014; 15:271. [PMID: 25112311 PMCID: PMC4267405 DOI: 10.1186/1471-2474-15-271] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2014] [Accepted: 06/28/2014] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Pulsed electromagnetic field (PEMF) is a non-invasive physical therapy used in the treatment of fracture nonunion or delayed healing. PEMF can facilitate the osteogenic differentiation of bone marrow mesenchymal stem cells in vitro. Amniotic epithelial cells (AECs) have been proposed as a potential source of stem cells for cell therapy. However, whether PEMF could modulate the osteogenic differentiation of AECs is unknown. In the present study, the effects of PEMF on the osteogenic differentiation of AECs were investigated. METHODS AECs were isolated from amniotic membrane of human placenta by trypsin digestion and were induced by PEMF and/or osteo-induction medium. After 21 days we used real time RT-PCR and immunocytochemistry to study the expression of osteoblast markers. The signal transduction of osteogenesis was further investigated. RESULTS The PEMF stimulation, or osteo-induction medium alone could induce osteogenic differentiation of AECs, as shown by expression of osteoblast specific genes and proteins including alkaline phosphatase and osteocalcin. Furthermore, a combination of PEMF and osteo-induction medium had synergy effects on osteogenic differentiation. In our study, the gene expression of BMP-2, Runx2, β-catenin, Nrf2, Keap1 and integrinβ1 were up-regulated in the osteogenic differentiation of AECs induced by PEMF and/or osteo-induction medium. CONCLUSIONS Combined application of PEMF and osteo-induction medium is synergistic for the osteogenic differentiation of AECs. It might be a novel approach in the bone regenerative medicine.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Xiaojing Liu
- Center of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, P, R, China.
| |
Collapse
|
33
|
Muehsam D, Ventura C. Life rhythm as a symphony of oscillatory patterns: electromagnetic energy and sound vibration modulates gene expression for biological signaling and healing. Glob Adv Health Med 2014; 3:40-55. [PMID: 24808981 PMCID: PMC4010966 DOI: 10.7453/gahmj.2014.008] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Affiliation(s)
- David Muehsam
- Visual Institute of Developmental Sciences, Bologna, Italy (Dr Muehsam)
| | - Carlo Ventura
- National Institute of Biostructures and Biosystems, Visual Institute of Developmental Sciences, Bologna; Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna (Dr Ventura), Italy
| |
Collapse
|
34
|
Liu DD, Ren Z, Yang G, Zhao QR, Mei YA. Melatonin protects rat cerebellar granule cells against electromagnetic field-induced increases in Na(+) currents through intracellular Ca(2+) release. J Cell Mol Med 2014; 18:1060-70. [PMID: 24548607 PMCID: PMC4508145 DOI: 10.1111/jcmm.12250] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2013] [Accepted: 01/18/2014] [Indexed: 01/12/2023] Open
Abstract
Although melatonin (MT) has been reported to protect cells against oxidative damage induced by electromagnetic radiation, few reports have addressed whether there are other protective mechanisms. Here, we investigated the effects of MT on extremely low-frequency electromagnetic field (ELF-EMF)-induced Nav activity in rat cerebellar granule cells (GCs). Exposing cerebellar GCs to ELF-EMF for 60 min. significantly increased the Nav current (INa ) densities by 62.5%. MT (5 μM) inhibited the ELF-EMF-induced INa increase. This inhibitory effect of MT is mimicked by an MT2 receptor agonist and was eliminated by an MT2 receptor antagonist. The Nav channel steady-state activation curve was significantly shifted towards hyperpolarization by ELF-EMF stimulation but remained unchanged by MT in cerebellar GC that were either exposed or not exposed to ELF-EMF. ELF-EMF exposure significantly increased the intracellular levels of phosphorylated PKA in cerebellar GCs, and both MT and IIK-7 did not reduce the ELF-EMF-induced increase in phosphorylated PKA. The inhibitory effects of MT on ELF-EMF-induced Nav activity was greatly reduced by the calmodulin inhibitor KN93. Calcium imaging showed that MT did not increase the basal intracellular Ca(2+) level, but it significantly elevated the intracellular Ca(2+) level evoked by the high K(+) stimulation in cerebellar GC that were either exposed or not exposed to ELF-EMF. In the presence of ruthenium red, a ryanodine-sensitive receptor blocker, the MT-induced increase in intracellular calcium levels was reduced. Our data show for the first time that MT protects against neuronal INa that result from ELF-EMF exposure through Ca(2+) influx-induced Ca(2+) release.
Collapse
Affiliation(s)
- Dong-Dong Liu
- School of Life Sciences, Institutes of Brain Science and State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, China
| | | | | | | | | |
Collapse
|
35
|
Bayır E, Bilgi E, Şendemir-Ürkmez A, Hameş-Kocabaş EE. The effects of different intensities, frequencies and exposure times of extremely low-frequency electromagnetic fields on the growth of Staphylococcus aureus and Escherichia coli O157:H7. Electromagn Biol Med 2013; 34:14-8. [PMID: 24279632 DOI: 10.3109/15368378.2013.853671] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
The impact of different types of extremely low-frequency electromagnetic fields (ELF-EMF) on the growth of Staphylococcus aureus and Escherichia coli O157:H7 was investigated. The cultures of bacteria in broth media were exposed to sinusoidal homogenous ELF-EMF with 2 and 4 mT magnetic intensities. Each intensity for each bacteria was combined with three different frequencies (20, 40 and 50 Hz), and four different exposure times (1, 2, 4 and 6 h). A cell suspension of each experiment was diluted for the appropriate range and inoculated to Mueller-Hinton Agar (MHA) plates after exposure to ELF-EMF. The number of colony forming units (CFU) of both strains was obtained after incubation at 37 °C for 24 h. Data were statistically evaluated by one-way analysis of variance (ANOVA), statistical significance was described at p < 0.05 and data were compared with their non-exposed controls. Magnetic intensity, frequency and exposure time of ELF-EMFs changed the characteristic responses for both microorganisms. Samples exposed to ELF-EMF showed a statistically significant decrease compared to their controls in colony forming capability, especially at long exposure times. An exposure to 4 mT-20 Hz ELF-EMF of 6 h produced maximum inhibition of CFU compared to their controls for both microorganisms (95.2% for S. aureus and 85% for E. coli).
Collapse
Affiliation(s)
- Ece Bayır
- Department of Biomedical Technologies, Graduate School of Natural and Applied Sciences and
| | | | | | | |
Collapse
|
36
|
Adipose-derived mesenchymal cells for bone regereneration: state of the art. BIOMED RESEARCH INTERNATIONAL 2013; 2013:416391. [PMID: 24307997 PMCID: PMC3838853 DOI: 10.1155/2013/416391] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Subscribe] [Scholar Register] [Received: 06/14/2013] [Accepted: 09/25/2013] [Indexed: 12/21/2022]
Abstract
Adipose tissue represents a hot topic in regenerative medicine because of the tissue source abundance, the relatively easy retrieval, and the inherent biological properties of mesenchymal stem cells residing in its stroma. Adipose-derived mesenchymal stem cells (ASCs) are indeed multipotent somatic stem cells exhibiting growth kinetics and plasticity, proved to induce efficient tissue regeneration in several biomedical applications. A defined consensus for their isolation, classification, and characterization has been very recently achieved. In particular, bone tissue reconstruction and regeneration based on ASCs has emerged as a promising approach to restore structure and function of bone compromised by injury or disease. ASCs have been used in combination with osteoinductive biomaterial and/or osteogenic molecules, in either static or dynamic culture systems, to improve bone regeneration in several animal models. To date, few clinical trials on ASC-based bone reconstruction have been concluded and proved effective. The aim of this review is to dissect the state of the art on ASC use in bone regenerative applications in the attempt to provide a comprehensive coverage of the topics, from the basic laboratory to recent clinical applications.
Collapse
|