1
|
Liao F, Li Y, Zhang Z, Yu Q, Liu H. Pulsed electromagnetic fields modulate energy metabolism during wound healing process: an in vitro model study. BMC Complement Med Ther 2025; 25:50. [PMID: 39939866 PMCID: PMC11823022 DOI: 10.1186/s12906-025-04792-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Accepted: 01/28/2025] [Indexed: 02/14/2025] Open
Abstract
BACKGROUND Pulsed electromagnetic fields (PEMFs) therapy was extensively investigated to treat wound healing, which is a highly metabolically demanding process. However, the effect of PEMFs on energy metabolism in wound healing remains largely unexplored. Therefore, our study aims to demonstrate the role of PEMFs on energy metabolism in wound healing. METHODS Scratch-wound healing assay and cell viability assay were performed for the in vitro study of the effect of PEMFs on cell migration and viability. Seahorse assay was conducted for energy metabolism analysis while holo-tomographic microscopy for fine changes of L929 cells. Mitochondrial membrane potential assay and intracellular reactive oxygen species (ROS) and pH assay were performed for analyzing the changes of mitochondrial function. RESULTS PEMFs with specific parameter (4mT, 80 Hz) promoted cell migration and viability. Glycolysis stress and mitochondria stress test revealed that PEMFs-exposed L929 cells was highly glycolytic for energy generation. Besides, PEMFs enhanced intracellular acidification and maintained low level of intracellular ROS in L929 cells. Compared to control group, much more vesicles were generated and then transported to regions close to the nuclear in L929 cells treated with PEMFs. CONCLUSIONS Our major findings revealed for the first time that PEMFs induce metabolic reprogramming of fibroblast shifting from mitochondrial respiration to glycolysis, accompanied with an increase of vesicular transport, which is closely related to wound healing in vitro.
Collapse
Affiliation(s)
- Feng Liao
- Department of Orthopedics, Sichuan Provincial People's Hospital,School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, PR China
| | - Yamei Li
- Department of Rehabilitation Medicine, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, PR China
| | - Zhou Zhang
- Department of Rehabilitation Medicine, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, PR China
| | - Qian Yu
- Department of Rehabilitation Medicine, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, PR China.
| | - Huifang Liu
- Department of Rehabilitation Medicine, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, PR China.
| |
Collapse
|
2
|
Liu Y, Zhao C, Zhang R, Pang Y, Li L, Feng S. Progression of mesenchymal stem cell regulation on imbalanced microenvironment after spinal cord injury. Stem Cell Res Ther 2024; 15:343. [PMID: 39354635 PMCID: PMC11446099 DOI: 10.1186/s13287-024-03914-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Accepted: 09/01/2024] [Indexed: 10/03/2024] Open
Abstract
Spinal cord injury (SCI) results in significant neural damage and inhibition of axonal regeneration due to an imbalanced microenvironment. Extensive evidence supports the efficacy of mesenchymal stem cell (MSC) transplantation as a therapeutic approach for SCI. This review aims to present an overview of MSC regulation on the imbalanced microenvironment following SCI, specifically focusing on inflammation, neurotrophy and axonal regeneration. The application, limitations and future prospects of MSC transplantation are discussed as well. Generally, a comprehensive perspective is provided for the clinical translation of MSC transplantation for SCI.
Collapse
Affiliation(s)
- Yifan Liu
- Department of Orthopaedics, Qilu Hospital of Shandong University, Shandong University Centre for Orthopaedics, Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, 107 West Wenhua Road, Lixia District, Jinan, 250012, Shandong, China
- Institute of Medical Sciences, The Second Hospital of Shandong University, Shandong University Center for Orthopaedics, Cheeloo College of Medicine, Shandong University, Jinan, 250033, Shandong, China
| | - Chenxi Zhao
- Department of Orthopaedics, Qilu Hospital of Shandong University, Shandong University Centre for Orthopaedics, Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, 107 West Wenhua Road, Lixia District, Jinan, 250012, Shandong, China
| | - Rong Zhang
- Department of Orthopaedics, Qilu Hospital of Shandong University, Shandong University Centre for Orthopaedics, Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, 107 West Wenhua Road, Lixia District, Jinan, 250012, Shandong, China
| | - Yilin Pang
- Department of Orthopaedics, Qilu Hospital of Shandong University, Shandong University Centre for Orthopaedics, Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, 107 West Wenhua Road, Lixia District, Jinan, 250012, Shandong, China
- Department of Orthopedics, International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin Key Laboratory of Spine and Spinal Cord Injury, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China
| | - Linquan Li
- Department of Orthopaedics, Qilu Hospital of Shandong University, Shandong University Centre for Orthopaedics, Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, 107 West Wenhua Road, Lixia District, Jinan, 250012, Shandong, China
| | - Shiqing Feng
- Department of Orthopaedics, Qilu Hospital of Shandong University, Shandong University Centre for Orthopaedics, Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, 107 West Wenhua Road, Lixia District, Jinan, 250012, Shandong, China.
- Institute of Medical Sciences, The Second Hospital of Shandong University, Shandong University Center for Orthopaedics, Cheeloo College of Medicine, Shandong University, Jinan, 250033, Shandong, China.
- Department of Orthopedics, International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin Key Laboratory of Spine and Spinal Cord Injury, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China.
| |
Collapse
|
3
|
Serrenho I, Ferreira SA, Baltazar G. Preconditioning of MSCs for Acute Neurological Conditions: From Cellular to Functional Impact-A Systematic Review. Cells 2024; 13:845. [PMID: 38786067 PMCID: PMC11119364 DOI: 10.3390/cells13100845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 05/02/2024] [Accepted: 05/14/2024] [Indexed: 05/25/2024] Open
Abstract
This systematic review aims to gather evidence on the mechanisms triggered by diverse preconditioning strategies for mesenchymal stem cells (MSCs) and their impact on their potential to treat ischemic and traumatic injuries affecting the nervous system. The 52 studies included in this review report nine different types of preconditioning, namely, manipulation of oxygen pressure, exposure to chemical substances, lesion mediators or inflammatory factors, usage of ultrasound, magnetic fields or biomechanical forces, and culture in scaffolds or 3D cultures. All these preconditioning strategies were reported to interfere with cellular pathways that influence MSCs' survival and migration, alter MSCs' phenotype, and modulate the secretome and proteome of these cells, among others. The effects on MSCs' phenotype and characteristics influenced MSCs' performance in models of injury, namely by increasing the homing and integration of the cells in the lesioned area and inducing the secretion of growth factors and cytokines. The administration of preconditioned MSCs promoted tissue regeneration, reduced neuroinflammation, and increased angiogenesis and myelinization in rodent models of stroke, traumatic brain injury, and spinal cord injury. These effects were also translated into improved cognitive and motor functions, suggesting an increased therapeutic potential of MSCs after preconditioning. Importantly, none of the studies reported adverse effects or less therapeutic potential with these strategies. Overall, we can conclude that all the preconditioning strategies included in this review can stimulate pathways that relate to the therapeutic effects of MSCs. Thus, it would be interesting to explore whether combining different preconditioning strategies can further boost the reparative effects of MSCs, solving some limitations of MSCs' therapy, namely donor-associated variability.
Collapse
Affiliation(s)
- Inês Serrenho
- Centro de Investigação em Ciências da Saúde (CICS-UBI), Universidade da Beira Interior, 6200-506 Covilhã, Portugal; (I.S.); (S.A.F.)
| | - Susana Alves Ferreira
- Centro de Investigação em Ciências da Saúde (CICS-UBI), Universidade da Beira Interior, 6200-506 Covilhã, Portugal; (I.S.); (S.A.F.)
| | - Graça Baltazar
- Faculdade de Ciências da Saúde, Universidade da Beira Interior, 6200-506 Covilhã, Portugal
| |
Collapse
|
4
|
Zhu LN, Chen D, He C. Metabolomics comparison of metabolites and functional pathways in the SH-SY5Y cell model of Parkinson's disease under PEMF exposure. Heliyon 2024; 10:e26540. [PMID: 38404789 PMCID: PMC10884933 DOI: 10.1016/j.heliyon.2024.e26540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2023] [Revised: 02/14/2024] [Accepted: 02/15/2024] [Indexed: 02/27/2024] Open
Abstract
Objective PEMF is an emerging technique in the treatment of Parkinson's disease (PD) due to its potential improvement of movement speed. The aim of this study was to investigate the metabolic profiles of pulsed electromagnetic fields (PEMFs) in an SH-SY5Y cell model of PD. Methods The SH-SY5Y cell model of PD was induced by 1-methyl-4-phenylpyridinium (MPP+). Liquid chromatography mass spectrometry (LC‒MS)-based untargeted metabolomics was performed to examine changes in the PD cell model with or without PEMF exposure. We conducted KEGG pathway enrichment analysis to explore the potentially related pathways of the differentially expressed metabolites. Results A total of 275 metabolites were annotated, and 27 significantly different metabolites were found between the PEMF treatment and control groups (VIP >1, P < 0.05), mainly including 4 amino acids and peptides, 4 fatty acid esters, 2 glycerophosphoethanolamines, 2 ceramides and 2 monoradylglycerols; among them, 12 metabolites were upregulated, and 15 were downregulated. The increased expression levels of glutamine, adenosine monophosphate and taurine were highly associated with PEMF stimulation in the PD model. The enrichment results of differentially abundant metabolite functional pathways showed that biological processes such as the mTOR signaling pathway, PI3K-Akt signaling pathway, and cAMP signaling pathway were significantly affected. Conclusion PEMFs affected glutamine, adenosine monophosphate and taurine as well as their functional pathways in an in vitro model of PD. Further functional studies regarding the biological effect of these changes are required to evaluate the clinical efficacy and safety of PEMF treatment in PD.
Collapse
Affiliation(s)
- Li-na Zhu
- Department of Rehabilitation Medicine, Key Laboratory of Rehabilitation Medicine, Institute of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Deng Chen
- Department of Neurology, West China Hospital, Sichuan University, Wai Nan Guo Xue Lane 37 #, Chengdu, 610041, Sichuan, China
| | - Chengqi He
- Department of Rehabilitation Medicine, Key Laboratory of Rehabilitation Medicine, Institute of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| |
Collapse
|
5
|
Cuenca-Ortolá I, Martínez-Rojas B, Moreno-Manzano V, García Castelló M, Monleón Pradas M, Martínez-Ramos C, Más Estellés J. A Strategy for Magnetic and Electric Stimulation to Enhance Proliferation and Differentiation of NPCs Seeded over PLA Electrospun Membranes. Biomedicines 2022; 10:2736. [PMID: 36359255 PMCID: PMC9687775 DOI: 10.3390/biomedicines10112736] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 10/07/2022] [Accepted: 10/25/2022] [Indexed: 09/30/2023] Open
Abstract
Neural progenitor cells (NPCs) have been shown to serve as an efficient therapeutic strategy in different cell therapy approaches, including spinal cord injury treatment. Despite the reported beneficial effects of NPC transplantation, the low survival and differentiation rates constrain important limitations. Herein, a new methodology has been developed to overcome both limitations by applying a combination of wireless electrical and magnetic stimulation to NPCs seeded on aligned poly(lactic acid) nanofibrous scaffolds for in vitro cell conditioning prior transplantation. Two stimulation patterns were tested and compared, continuous (long stimulus applied once a day) and intermittent (short stimulus applied three times a day). The results show that applied continuous stimulation promotes NPC proliferation and preferential differentiation into oligodendrocytic and neuronal lineages. A neural-like phenotypic induction was observed when compared to unstimulated NPCs. In contrast, intermittent stimulation patterns did not affect NPC proliferation and differentiation to oligodendrocytes or astrocytes morphology with a detrimental effect on neuronal differentiation. This study provides a new approach of using a combination of electric and magnetic stimulation to induce proliferation and further neuronal differentiation, which would improve therapy outcomes in disorders such as spinal cord injury.
Collapse
Affiliation(s)
- Irene Cuenca-Ortolá
- Center for Biomaterials and Tissue Engineering, Universitat Politècnica de València, Cno. de Vera s/n, 46022 Valencia, Spain
| | - Beatriz Martínez-Rojas
- Neuronal and Tissue Regeneration Laboratory, Centro de Investigación Príncipe Felipe, 46012 Valencia, Spain
| | - Victoria Moreno-Manzano
- Neuronal and Tissue Regeneration Laboratory, Centro de Investigación Príncipe Felipe, 46012 Valencia, Spain
| | - Marcos García Castelló
- Center for Biomaterials and Tissue Engineering, Universitat Politècnica de València, Cno. de Vera s/n, 46022 Valencia, Spain
| | - Manuel Monleón Pradas
- Center for Biomaterials and Tissue Engineering, Universitat Politècnica de València, Cno. de Vera s/n, 46022 Valencia, Spain
- Biomedical Research Networking Center in Bioengineering Biomaterials and Nanomedicine, CIBER-BBN, 28029 Madrid, Spain
| | - Cristina Martínez-Ramos
- Center for Biomaterials and Tissue Engineering, Universitat Politècnica de València, Cno. de Vera s/n, 46022 Valencia, Spain
- Unitat Predepartamental de Medicina, Universitat Jaume I, Avda/Sos Baynat, s/n, 12071 Castellón de la Plana, Spain
| | - Jorge Más Estellés
- Center for Biomaterials and Tissue Engineering, Universitat Politècnica de València, Cno. de Vera s/n, 46022 Valencia, Spain
| |
Collapse
|
6
|
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
|
7
|
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
|
8
|
Li M, Jiang Y, Hou Q, Zhao Y, Zhong L, Fu X. Potential pre-activation strategies for improving therapeutic efficacy of mesenchymal stem cells: current status and future prospects. Stem Cell Res Ther 2022; 13:146. [PMID: 35379361 PMCID: PMC8981790 DOI: 10.1186/s13287-022-02822-2] [Citation(s) in RCA: 64] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 03/20/2022] [Indexed: 12/13/2022] Open
Abstract
Mesenchymal stem cell (MSC)-based therapy has been considered as a promising approach targeting a variety of intractable diseases due to remarkable multiple effect of MSCs, such as multilineage differentiation, immunomodulatory property, and pro-regenerative capacity. However, poor engraftment, low survival rate of transplanted MSC, and impaired donor-MSC potency under host age/disease result in unsatisfactory therapeutic outcomes. Enhancement strategies, including genetic manipulation, pre-activation, and modification of culture method, have been investigated to generate highly functional MSC, and approaches for MSC pre-activation are highlighted. In this review, we summarized the current approaches of MSC pre-activation and further classified, analysed the scientific principles and main characteristics of these manipulations, and described the pros and cons of individual pre-activation strategies. We also discuss the specialized tactics to solve the challenges in this promising field so that it improves MSC therapeutic functions to serve patients better.
Collapse
Affiliation(s)
- Meirong Li
- Research Center for Tissue Repair and Regeneration Affiliated to the Medical Innovation Research Division and 4th Medical Center, PLA General Hospital and PLA Medical College, Beijing, China. .,PLA Key Laboratory of Tissue Repair and Regenerative Medicine and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration, Beijing, China. .,Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences 2019RU051, Beijing, China.
| | - Yufeng Jiang
- Wound Repairing Department, PLA Strategic Support Force Characteristic Medical Center, Beijing, 100101, China
| | - Qian Hou
- Research Center for Tissue Repair and Regeneration Affiliated to the Medical Innovation Research Division and 4th Medical Center, PLA General Hospital and PLA Medical College, Beijing, China.,PLA Key Laboratory of Tissue Repair and Regenerative Medicine and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration, Beijing, China.,Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences 2019RU051, Beijing, China
| | - Yali Zhao
- Central Laboratory, Trauma Treatment Center, Chinese PLA General Hospital, Hainan Hospital, Sanya, China
| | - Lingzhi Zhong
- Research Center for Tissue Repair and Regeneration Affiliated to the Medical Innovation Research Division and 4th Medical Center, PLA General Hospital and PLA Medical College, Beijing, China.,PLA Key Laboratory of Tissue Repair and Regenerative Medicine and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration, Beijing, China.,Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences 2019RU051, Beijing, China
| | - Xiaobing Fu
- Research Center for Tissue Repair and Regeneration Affiliated to the Medical Innovation Research Division and 4th Medical Center, PLA General Hospital and PLA Medical College, Beijing, China. .,PLA Key Laboratory of Tissue Repair and Regenerative Medicine and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration, Beijing, China. .,Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences 2019RU051, Beijing, China.
| |
Collapse
|
9
|
Circadian Rhythm Modulates the Therapeutic Activity of Pulsed Electromagnetic Fields on Intervertebral Disc Degeneration in Rats. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:9067611. [PMID: 35368872 PMCID: PMC8975688 DOI: 10.1155/2022/9067611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 02/23/2022] [Accepted: 03/10/2022] [Indexed: 11/29/2022]
Abstract
Circadian rhythm (CR) imparts significant benefits in treating multiple diseases, such as heart diseases and arthritis. But the CR effect on intervertebral disc degeneration (IVDD) therapy remains unclear. Recent studies revealed that pulsed electromagnetic fields (PEMF) are capable of alleviating IVDD. In this study, we evaluated the CR-mediated regulation of PEMF therapeutic effect on IVDD induced by rat tail disc needle puncture. Our results demonstrated that the daytime PEMF stimulation (DPEMF) is more effective than the nighttime PEMF (NPEMF) in delaying IVDD. Moreover, the rats treated with DPEMF maintained better disc stability and histology after 8 weeks, relative to NPEMF. CR and PEMF cotherapies were also examined in cellular models, whereby serum shock was used to induce different levels of clock gene expression in the nucleus pulposus (NP), thus imitating CR in vitro. PEMF at ZT8 (higher level of clock gene expression) correlated with a higher extracellular matrix (ECM) component expression, compared to ZT20 (lower level of clock gene expression). Taken together, these data suggest a strong role of CR in regulating the beneficial effect of PEMF on IVDD. Our findings provide a potential clinical significance of CR in optimizing PEMF positive effects on IVDD.
Collapse
|
10
|
Romeo S, Zeni O, Scarfì MR, Poeta L, Lioi MB, Sannino A. Radiofrequency Electromagnetic Field Exposure and Apoptosis: A Scoping Review of In Vitro Studies on Mammalian Cells. Int J Mol Sci 2022; 23:2322. [PMID: 35216437 PMCID: PMC8877695 DOI: 10.3390/ijms23042322] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 02/11/2022] [Accepted: 02/16/2022] [Indexed: 02/01/2023] Open
Abstract
In the last decades, experimental studies have been carried out to investigate the effects of radiofrequency (RF, 100 kHz-300 GHz) electromagnetic fields (EMF) exposure on the apoptotic process. As evidence-based critical evaluation of RF and apoptosis in vitro is lacking, we performed a scoping literature review with the aim of systematically mapping the research performed in this area and identifying gaps in knowledge. Eligible for inclusion were in vitro studies assessing apoptosis in mammalian cells exposed to RF-EMF, which met basic quality criteria (sham control, at least three independent experiments, appropriate dosimetry analysis and temperature monitoring). We conducted a systematic literature review and charted data in order to overview the main characteristics of included studies. From the 4362 papers retrieved with our search strategy, 121 were pertinent but, among them, only 42 met basic quality criteria. We pooled data with respect to exposure (frequency, exposure level and duration) and biological parameters (cell type, endpoint), and highlighted some qualitative trends with respect to the detection of significant effect of RF-EMF on the apoptotic process. We provided a qualitative picture of the evidence accumulated so far, and highlighted that the quality of experimental methodology still needs to be highly improved.
Collapse
Affiliation(s)
- Stefania Romeo
- CNR, Institute for Electromagnetic Sensing of the Environment (IREA) via Diocleziano 328, 80124 Napoli, Italy; (S.R.); (M.R.S.); (L.P.); (M.B.L.); (A.S.)
| | - Olga Zeni
- CNR, Institute for Electromagnetic Sensing of the Environment (IREA) via Diocleziano 328, 80124 Napoli, Italy; (S.R.); (M.R.S.); (L.P.); (M.B.L.); (A.S.)
| | - Maria Rosaria Scarfì
- CNR, Institute for Electromagnetic Sensing of the Environment (IREA) via Diocleziano 328, 80124 Napoli, Italy; (S.R.); (M.R.S.); (L.P.); (M.B.L.); (A.S.)
| | - Loredana Poeta
- CNR, Institute for Electromagnetic Sensing of the Environment (IREA) via Diocleziano 328, 80124 Napoli, Italy; (S.R.); (M.R.S.); (L.P.); (M.B.L.); (A.S.)
| | - Maria Brigida Lioi
- CNR, Institute for Electromagnetic Sensing of the Environment (IREA) via Diocleziano 328, 80124 Napoli, Italy; (S.R.); (M.R.S.); (L.P.); (M.B.L.); (A.S.)
- Department of Science, University of Basilicata, Viale dell’Ateneo Lucano, 85100 Potenza, Italy
| | - Anna Sannino
- CNR, Institute for Electromagnetic Sensing of the Environment (IREA) via Diocleziano 328, 80124 Napoli, Italy; (S.R.); (M.R.S.); (L.P.); (M.B.L.); (A.S.)
| |
Collapse
|
11
|
Cho H, Park HJ, Seo YK. Induction of PLXNA4 Gene during Neural Differentiation in Human Umbilical-Cord-Derived Mesenchymal Stem Cells by Low-Intensity Sub-Sonic Vibration. Int J Mol Sci 2022; 23:ijms23031522. [PMID: 35163445 PMCID: PMC8835879 DOI: 10.3390/ijms23031522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Revised: 01/22/2022] [Accepted: 01/27/2022] [Indexed: 02/01/2023] Open
Abstract
Human umbilical-cord-derived mesenchymal stem cells (hUC-MSC) are a type of mesenchymal stem cells and are more primitive than other MSCs. In this study, we identify novel genes and signal-activating proteins involved in the neural differentiation of hUC-MSCs induced by Low-Intensity Sub-Sonic Vibration (LISSV). RNA sequencing was used to find genes involved in the differentiation process by LISSV. The changes in hUC-MSCs caused by LISSV were confirmed by PLXNA4 overexpression and gene knockdown through small interfering RNA experiments. The six genes were increased among genes related to neurons and the nervous system. One of them, the PLXNA4 gene, is known to play a role as a guide for axons in the development of the nervous system. When the PLXNA4 recombinant protein was added, neuron-related genes were increased. In the PLXNA4 gene knockdown experiment, the expression of neuron-related genes was not changed by LISSV exposure. The PLXNA4 gene is activated by sema family ligands. The expression of SEMA3A was increased by LISSV, and its downstream signaling molecule, FYN, was also activated. We suggest that the PLXNA4 gene plays an important role in hUC-MSC neuronal differentiation through exposure to LISSV. The differentiation process depends on SEMA3A-PLXNA4-dependent FYN activation in hUC-MSCs.
Collapse
Affiliation(s)
- Hyunjin Cho
- Research Institute of Integrative Life Sciences, Dongguk University, Goyang-si 10326, Korea;
| | - Hee-Jung Park
- Department of Medical Biotechnology (BK21 Plus Team), Dongguk University, Goyang-si 10326, Korea;
| | - Young-Kwon Seo
- Department of Medical Biotechnology (BK21 Plus Team), Dongguk University, Goyang-si 10326, Korea;
- Correspondence:
| |
Collapse
|
12
|
Hu XM, Zhang Q, Zhou RX, Wu YL, Li ZX, Zhang DY, Yang YC, Yang RH, Hu YJ, Xiong K. Programmed cell death in stem cell-based therapy: Mechanisms and clinical applications. World J Stem Cells 2021; 13:386-415. [PMID: 34136072 PMCID: PMC8176847 DOI: 10.4252/wjsc.v13.i5.386] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 04/26/2021] [Accepted: 05/07/2021] [Indexed: 02/06/2023] Open
Abstract
Stem cell-based therapy raises hopes for a better approach to promoting tissue repair and functional recovery. However, transplanted stem cells show a high death percentage, creating challenges to successful transplantation and prognosis. Thus, it is necessary to investigate the mechanisms underlying stem cell death, such as apoptotic cascade activation, excessive autophagy, inflammatory response, reactive oxygen species, excitotoxicity, and ischemia/hypoxia. Targeting the molecular pathways involved may be an efficient strategy to enhance stem cell viability and maximize transplantation success. Notably, a more complex network of cell death receives more attention than one crucial pathway in determining stem cell fate, highlighting the challenges in exploring mechanisms and therapeutic targets. In this review, we focus on programmed cell death in transplanted stem cells. We also discuss some promising strategies and challenges in promoting survival for further study.
Collapse
Affiliation(s)
- Xi-Min Hu
- Department of Anatomy and Neurobiology, School of Basic Medical Sciences, Central South University, Changsha 410013, Hunan Province, China
| | - Qi Zhang
- Department of Anatomy and Neurobiology, School of Basic Medical Sciences, Central South University, Changsha 410013, Hunan Province, China
| | - Rui-Xin Zhou
- Department of Anatomy and Neurobiology, School of Basic Medical Sciences, Central South University, Changsha 410013, Hunan Province, China
| | - Yan-Lin Wu
- Department of Anatomy and Neurobiology, School of Basic Medical Sciences, Central South University, Changsha 410013, Hunan Province, China
| | - Zhi-Xin Li
- Department of Anatomy and Neurobiology, School of Basic Medical Sciences, Central South University, Changsha 410013, Hunan Province, China
| | - Dan-Yi Zhang
- Department of Anatomy and Neurobiology, School of Basic Medical Sciences, Central South University, Changsha 410013, Hunan Province, China
| | - Yi-Chao Yang
- Department of Anatomy and Neurobiology, School of Basic Medical Sciences, Central South University, Changsha 410013, Hunan Province, China
| | - Rong-Hua Yang
- Department of Burns, Fo Shan Hospital of Sun Yat-Sen University, Foshan 528000, Guangdong Province, China
| | - Yong-Jun Hu
- Department of Cardiovascular Medicine, Hunan People's Hospital (the First Affiliated Hospital of Hunan Normal University, Changsha 410005, Hunan Province, China
| | - Kun Xiong
- Department of Anatomy and Neurobiology, School of Basic Medical Sciences, Central South University, Changsha 410013, Hunan Province, China.
| |
Collapse
|
13
|
Long-term treatment with transcranial pulsed electromagnetic fields improves movement speed and elevates cerebrospinal erythropoietin in Parkinson's disease. PLoS One 2021; 16:e0248800. [PMID: 33909634 PMCID: PMC8081215 DOI: 10.1371/journal.pone.0248800] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2020] [Accepted: 03/04/2021] [Indexed: 12/01/2022] Open
Abstract
Background Parkinson’s disease is characterized by motor dysfunctions including bradykinesia. In a recent study, eight weeks of daily transcranial stimulation with bipolar pulsed electromagnetic fields improved functional rate of force development and decreased inter-hand tremor coherence in patients with mild Parkinson’s disease. Objective To investigate the effect of long-term treatment with transcranial bipolar pulsed electromagnetic fields on motor performance in terms of movement speed and on neurotrophic and angiogenic factors. Methods Patients diagnosed with idiopathic Parkinson’s disease had either daily 30-min treatment with bipolar (±50 V) transcranial pulsed electromagnetic stimulation (squared pulses, 3ms duration) for three eight-week periods separated by one-week pauses (T-PEMF group) (n = 16) or were included in a PD-control group (n = 8). Movement speed was assessed in a six-cycle sit-to-stand task performed on a force plate. Cerebrospinal fluid and venous blood were collected and analyzed for erythropoietin and vascular endothelial growth factor. Results Major significant improvement of movement speed compared to the natural development of the disease was found (p = 0.001). Thus, task completion time decreased gradually during the treatment period from 10.10s to 8.23s (p<0.001). The untreated PD-control group did not change (p = 0.458). The treated group did not differ statistically from that of a healthy age matched reference group at completion of treatment. Erythropoietin concentration in the cerebrospinal fluid also increased significantly in the treated group (p = 0.012). Conclusion Long-term treatment with transcranial bipolar pulsed electromagnetic fields increased movement speed markedly and elevated erythropoietin levels. We hypothesize that treatment with transcranial bipolar pulsed electromagnetic fields improved functional performance by increasing dopamine levels in the brain, possibly through erythropoietin induced neural repair and/or protection of dopaminergic neurons.
Collapse
|
14
|
Sim WS, Park BW, Ban K, Park HJ. In Situ Preconditioning of Human Mesenchymal Stem Cells Elicits Comprehensive Cardiac Repair Following Myocardial Infarction. Int J Mol Sci 2021; 22:1449. [PMID: 33535594 PMCID: PMC7867207 DOI: 10.3390/ijms22031449] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2021] [Revised: 01/27/2021] [Accepted: 01/28/2021] [Indexed: 01/04/2023] Open
Abstract
Human bone marrow-derived mesenchymal stem cells (BM-MSCs), represented as a population of adult stem cells, have long been considered as one of the most promising sources for cell-based cardiac regenerative therapy. However, their clinical use has been significantly hampered by low survival and poor retention following administration into failing hearts. Here, to improve the therapeutic effectiveness of BM-MSCs, we examined a novel therapeutic platform named in situ preconditioning in a rat myocardial infarction (MI) model. In situ preconditioning was induced by a combinatory treatment of BM-MSCs with genetically engineered hepatocyte growth factor-expressing MSCs (HGF-eMSCs) and heart-derived extracellular matrix (hdECM) hydrogel. Subsequently, our results demonstrated that in situ preconditioning with cell mixture substantially improved the survival/retention of BM-MSCs in the MI-induced rat hearts. Enhanced retention of BM-MSCs ultimately led to a significant cardiac function improvement, which was derived from the protection of myocardium and enhancement of vessel formation in the MI hearts. The results provide compelling evidence that in situ preconditioning devised to improve the therapeutic potential of BM-MSCs can be an effective strategy to achieve cardiac repair of MI hearts.
Collapse
Affiliation(s)
- Woo-Sup Sim
- Department of Biomedicine & Health Sciences, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul 137701, Korea; (W.-S.S.); (B.-W.P.)
- Division of Cardiology, Department of Internal Medicine, Seoul St. Mary’s Hospital, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul 137701, Korea
| | - Bong-Woo Park
- Department of Biomedicine & Health Sciences, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul 137701, Korea; (W.-S.S.); (B.-W.P.)
- Division of Cardiology, Department of Internal Medicine, Seoul St. Mary’s Hospital, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul 137701, Korea
| | - Kiwon Ban
- Department of Biomedical Sciences, City University of Hong Kong, Tat Chee Avenue, Kowloon, Hong Kong, China
| | - Hun-Jun Park
- Department of Biomedicine & Health Sciences, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul 137701, Korea; (W.-S.S.); (B.-W.P.)
- Division of Cardiology, Department of Internal Medicine, Seoul St. Mary’s Hospital, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul 137701, Korea
- Cell Death Disease Research Center, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea
| |
Collapse
|
15
|
Bui TVA, Hwang JW, Lee JH, Park HJ, Ban K. Challenges and Limitations of Strategies to Promote Therapeutic Potential of Human Mesenchymal Stem Cells for Cell-Based Cardiac Repair. Korean Circ J 2021; 51:97-113. [PMID: 33525065 PMCID: PMC7853896 DOI: 10.4070/kcj.2020.0518] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Accepted: 12/16/2020] [Indexed: 12/18/2022] Open
Abstract
Mesenchymal stem cells (MSCs) represent a population of adult stem cells residing in many tissues, mainly bone marrow, adipose tissue, and umbilical cord. Due to the safety and availability of standard procedures and protocols for isolation, culturing, and characterization of these cells, MSCs have emerged as one of the most promising sources for cell-based cardiac regenerative therapy. Once transplanted into a damaged heart, MSCs release paracrine factors that nurture the injured area, prevent further adverse cardiac remodeling, and mediate tissue repair along with vasculature. Numerous preclinical studies applying MSCs have provided significant benefits following myocardial infarction. Despite promising results from preclinical studies using animal models, MSCs are not up to the mark for human clinical trials. As a result, various approaches have been considered to promote the therapeutic potency of MSCs, such as genetic engineering, physical treatments, growth factor, and pharmacological agents. Each strategy has targeted one or multi-potentials of MSCs. In this review, we will describe diverse approaches that have been developed to promote the therapeutic potential of MSCs for cardiac regenerative therapy. Particularly, we will discuss major characteristics of individual strategy to enhance therapeutic efficacy of MSCs including scientific principles, advantages, limitations, and improving factors. This article also will briefly introduce recent novel approaches that MSCs enhanced therapeutic potentials of other cells for cardiac repair.
Collapse
Affiliation(s)
- Thi Van Anh Bui
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong SAR, China
| | - Ji Won Hwang
- Department of Biomedicine & Health Sciences, The Catholic University of Korea, Seoul, Korea.,Division of Cardiology, Department of Internal Medicine, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, Korea
| | - Jung Hoon Lee
- Department of Chemistry, City University of Hong Kong, Hong Kong SAR, China
| | - Hun Jun Park
- Department of Biomedicine & Health Sciences, The Catholic University of Korea, Seoul, Korea.,Division of Cardiology, Department of Internal Medicine, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, Korea.,Cell Death Disease Research Center, College of Medicine, The Catholic University of Korea, Seoul, Korea.
| | - Kiwon Ban
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong SAR, China.
| |
Collapse
|
16
|
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
|
17
|
Miller CP, Prener M, Dissing S, Paulson OB. Transcranial low-frequency pulsating electromagnetic fields (T-PEMF) as post-concussion syndrome treatment. Acta Neurol Scand 2020; 142:597-604. [PMID: 32559313 PMCID: PMC7689717 DOI: 10.1111/ane.13300] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 05/31/2020] [Accepted: 06/15/2020] [Indexed: 01/01/2023]
Abstract
Background Treatment options for the subgroup of people who develop long‐lasting symptoms following mild traumatic brain injury are limited. Transcranial pulsating low‐frequency electromagnetic stimulation (T‐PEMF) in other patient groups has shown promising results in several studies with proposed neuroprotective and anti‐inflammatory effects. Objective The present pilot study was conducted to access feasibility and tolerability of T‐PEMF in treating post‐concussion syndrome. Methods Seven patients with post‐concussion syndrome received 5 weeks of daily 30 minutes T‐PEMF treatment with evaluation after 2 and 5 weeks and 3 months after ending treatment. Results Compliance was high as all subject completed the full treatment. Two patients however experienced a worsening of their concussion symptoms during the course of treatment. The remaining patients had some discomfort in relation to treatment, mainly headache, but passing and less for each treatment. The majority (n = 5) had a reduction in symptoms overall, up to 61% (2%‐61%) based on the Rivermead Post‐Concussion Symptoms Questionnaire. Conclusion Further studies on T‐PEMF as a treatment option for post‐concussion syndrome are warranted.
Collapse
Affiliation(s)
- Claire Prener Miller
- Neurobiology Research Unit Department of Neurology Rigshospitalet Copenhagen Denmark
| | - Martin Prener
- Neurobiology Research Unit Department of Neurology Rigshospitalet Copenhagen Denmark
- Department of Clinical Medicine University of Copenhagen Copenhagen Denmark
| | - Steen Dissing
- Department of Cellular and Molecular Medicine Panum Institute University of Copenhagen Copenhagen Denmark
| | - Olaf B. Paulson
- Neurobiology Research Unit Department of Neurology Rigshospitalet Copenhagen Denmark
- Department of Clinical Medicine University of Copenhagen Copenhagen Denmark
| |
Collapse
|
18
|
Choudhary P, Gupta A, Singh S. Therapeutic Advancement in Neuronal Transdifferentiation of Mesenchymal Stromal Cells for Neurological Disorders. J Mol Neurosci 2020; 71:889-901. [PMID: 33047251 DOI: 10.1007/s12031-020-01714-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Accepted: 09/16/2020] [Indexed: 12/12/2022]
Abstract
Neurodegenerative disorders have become the leading cause of chronic pain and death. Treatments available are not sufficient to help the patients as they only alleviate the symptoms and not the cause. In this regard, stem cells therapy has emerged as an upcoming option for the replacement of dead and damaged neurons. Stem cells, in general, are characterized as cells exhibiting potency properties, i.e., on being subjected to specific conditions they transform into cells of another lineage. Of all the types, mesenchymal stem cells (MSCs) are known for their pluripotent nature without the obstacle of ethical concern surrounding the procurement of other cell types. Although fibroblasts are quite similar to MSCs morphologically, certain markers like CD73, CD 90 are specific to MSCs, making both the cell types distinguishable from each other. This is implemented while procuring MSCs from a plethora of sources like umbilical cord blood, adipose tissue, bone marrow, etc. Among these, bone marrow MSCs are the most widely used type for neural regeneration. Neural regeneration is achieved via transdifferentiation. Several studies have either transplanted the stem cells into rodent models or have carried out transdifferentiation in vitro. The process involves a combination of growth factors, pre-treatment factors, and neuronal differentiation inducing mediums. The results obtained are characterized by neuron-like morphology, expression of markers, along with electrophysical activity in some. Recent attempts involve exploring biomaterials that may mimic the native ECM and therefore can be directly introduced at the site of interest. The review gives a brief description of MSCs, their sources and markers, and the different attempts that have been made towards achieving the goal of differentiating MSCs into neurons.
Collapse
Affiliation(s)
- Princy Choudhary
- Applied Science Department, Indian Institute of Information Technology, Allahabad, UP, India
| | - Ayushi Gupta
- Applied Science Department, Indian Institute of Information Technology, Allahabad, UP, India
| | - Sangeeta Singh
- Applied Science Department, Indian Institute of Information Technology, Allahabad, UP, India.
| |
Collapse
|
19
|
Grasso R, Pellitteri R, Caravella SA, Musumeci F, Raciti G, Scordino A, Sposito G, Triglia A, Campisi A. Dynamic changes in cytoskeleton proteins of olfactory ensheathing cells induced by radiofrequency electromagnetic fields. J Exp Biol 2020; 223:jeb217190. [PMID: 32041804 DOI: 10.1242/jeb.217190] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Accepted: 02/02/2020] [Indexed: 01/13/2023]
Abstract
Several evidences have suggested the ability of radiofrequency electromagnetic fields to influence biological systems, even if the action mechanisms are not well understood. There are few data on the effect of radiofrequency electromagnetic fields on self-renewal of neural progenitor cells. A particular glial type that shows characteristics of stem cells is olfactory ensheathing cells (OECs). Herein, we assessed the non-thermal effects induced on OECs through radiofrequency electromagnetic fields changing the envelope of the electromagnetic wave. Primary OEC cultures were exposed to continuous or amplitude-modulated 900 MHz electromagnetic fields, in the far-field condition and at different exposure times (10, 15, 20 min). The expression of OEC markers (S-100 and nestin), cytoskeletal proteins (GFAP and vimentin), apoptotic pathway activation by caspase-3 cleavage and cell viability were evaluated. Our results highlight that 20 min of exposure to continuous or amplitude-modulated 900 MHz electromagnetic fields induced a different and significant decrease in cell viability. In addition, according to the electromagnetic field waveform, diverse dynamic changes in the expression of the analysed markers in OECs and activation of the apoptotic pathway were observed. The data suggest that radiofrequency electromagnetic fields might play different and important roles in the self-renewal of OEC stem cells, which are involved in nervous system repair.
Collapse
Affiliation(s)
- Rosaria Grasso
- Department of Physics and Astronomy 'Ettore Majorana', University of Catania, 95123 Catania, Italy
- Laboratori Nazionali del Sud, National Institute for Nuclear Physics, 95123 Catania, Italy
| | - Rosalia Pellitteri
- Institute for Biomedical Research and Innovation, Italian National Research Council, 95126 Catania, Italy
| | | | - Francesco Musumeci
- Department of Physics and Astronomy 'Ettore Majorana', University of Catania, 95123 Catania, Italy
- Laboratori Nazionali del Sud, National Institute for Nuclear Physics, 95123 Catania, Italy
| | - Giuseppina Raciti
- Department of Drug Sciences, Section of Biochemistry, University of Catania, 95125 Catania, Italy
| | - Agata Scordino
- Department of Physics and Astronomy 'Ettore Majorana', University of Catania, 95123 Catania, Italy
- Laboratori Nazionali del Sud, National Institute for Nuclear Physics, 95123 Catania, Italy
| | - Giovanni Sposito
- Department of Drug Sciences, Section of Biochemistry, University of Catania, 95125 Catania, Italy
| | - Antonio Triglia
- Department of Physics and Astronomy 'Ettore Majorana', University of Catania, 95123 Catania, Italy
| | - Agata Campisi
- Department of Drug Sciences, Section of Biochemistry, University of Catania, 95125 Catania, Italy
| |
Collapse
|
20
|
Optical approaches for single-cell and subcellular analysis of GPCR-G protein signaling. Anal Bioanal Chem 2019; 411:4481-4508. [PMID: 30927013 DOI: 10.1007/s00216-019-01774-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2018] [Revised: 03/05/2019] [Accepted: 03/08/2019] [Indexed: 01/05/2023]
Abstract
G protein-coupled receptors (GPCRs), G proteins, and their signaling associates are major signal transducers that control the majority of cellular signaling and regulate key biological functions including immune, neurological, cardiovascular, and metabolic processes. These pathways are targeted by over one-third of drugs on the market; however, the current understanding of their function is limited and primarily derived from cell-destructive approaches providing an ensemble of static, multi-cell information about the status and composition of molecules. Spatiotemporal behavior of molecules involved is crucial to understanding in vivo cell behaviors both in health and disease, and the advent of genetically encoded fluorescence proteins and small fluorophore-based biosensors has facilitated the mapping of dynamic signaling in cells with subcellular acuity. Since we and others have developed optogenetic methods to regulate GPCR-G protein signaling in single cells and subcellular regions using dedicated wavelengths, the desire to develop and adopt optogenetically amenable assays to measure signaling has motivated us to take a broader look at the available optical tools and approaches compatible with measuring single-cell and subcellular GPCR-G protein signaling. Here we review such key optical approaches enabling the examination of GPCR, G protein, secondary messenger, and downstream molecules such as kinase and lipid signaling in living cells. The methods reviewed employ both fluorescence and bioluminescence detection. We not only further elaborate the underlying principles of these sensors but also discuss the experimental criteria and limitations to be considered during their use in single-cell and subcellular signal mapping.
Collapse
|
21
|
Ross CL, Ang DC, Almeida-Porada G. Targeting Mesenchymal Stromal Cells/Pericytes (MSCs) With Pulsed Electromagnetic Field (PEMF) Has the Potential to Treat Rheumatoid Arthritis. Front Immunol 2019; 10:266. [PMID: 30886614 PMCID: PMC6409305 DOI: 10.3389/fimmu.2019.00266] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Accepted: 01/31/2019] [Indexed: 01/14/2023] Open
Abstract
Rheumatoid arthritis (RA) is a systemic autoimmune disease characterized by chronic inflammation of synovium (synovitis), with inflammatory/immune cells and resident fibroblast-like synoviocytes (FLS) acting as major players in the pathogenesis of this disease. The resulting inflammatory response poses considerable risks as loss of bone and cartilage progresses, destroying the joint surface, causing joint damage, joint failure, articular dysfunction, and pre-mature death if left untreated. At the cellular level, early changes in RA synovium include inflammatory cell infiltration, synovial hyperplasia, and stimulation of angiogenesis to the site of injury. Different angiogenic factors promote this disease, making the role of anti-angiogenic therapy a focus of RA treatment. To control angiogenesis, mesenchymal stromal cells/pericytes (MSCs) in synovial tissue play a vital role in tissue repair. While recent evidence reports that MSCs found in joint tissues can differentiate to repair damaged tissue, this repair function can be repressed by the inflammatory milieu. Extremely-low frequency pulsed electromagnetic field (PEMF), a biophysical form of stimulation, has an anti-inflammatory effect by causing differentiation of MSCs. PEMF has also been reported to increase the functional activity of MSCs to improve differentiation to chondrocytes and osteocytes. Moreover, PEMF has been demonstrated to accelerate cell differentiation, increase deposition of collagen, and potentially return vascular dysfunction back to homeostasis. The aim of this report is to review the effects of PEMF on MSC modulation of cytokines, growth factors, and angiogenesis, and describe its effect on MSC regeneration of synovial tissue to further understand its potential role in the treatment of RA.
Collapse
Affiliation(s)
- Christina L Ross
- Wake Forest Institute for Regenerative Medicine, Winston-Salem, NC, United States.,Wake Forest Center for Integrative Medicine, Wake Forest School of Medicine, Winston-Salem, NC, United States
| | - Dennis C Ang
- Department of Rheumatology and Immunology, Wake Forest School of Medicine, Winston-Salem, NC, United States
| | - Graça Almeida-Porada
- Wake Forest Institute for Regenerative Medicine, Winston-Salem, NC, United States
| |
Collapse
|
22
|
Malling ASB, Morberg BM, Wermuth L, Gredal O, Bech P, Jensen BR. The effect of 8 weeks of treatment with transcranial pulsed electromagnetic fields on hand tremor and inter-hand coherence in persons with Parkinson's disease. J Neuroeng Rehabil 2019; 16:19. [PMID: 30704504 PMCID: PMC6357382 DOI: 10.1186/s12984-019-0491-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Accepted: 01/23/2019] [Indexed: 12/05/2022] Open
Abstract
BACKGROUND Parkinson's disease (PD) tremor comprises asymmetric rest and postural tremor with unilateral onset. Tremor intensity can be amplified by stress and reduced by attention, and the medical treatment is complex. Mirror movements and unintentional synchronization of bimanual movements, possibly caused by insufficient inhibition of inter-hemispheric crosstalk, have been reported in PD, indicating a lag of lateralization. Potential neuroprotective effects of pulsed electromagnetic fields (PEMF) have been reported in-vitro and in rodents, as have influences of PEMF on human tremor. The aim was to investigate the effect of 8 weeks daily transcranial PEMF treatment (T-PEMF) of persons with PD on rest and postural hand tremor characteristics and on inter-hand coherence. METHODS Hand accelerations of 50 PD participants with uni- or bilateral tremor participating in a clinical trial were analysed. A rest and postural tremor task performed during serial subtraction was assessed before and after 8 weeks of T-PEMF (30 min/day, 50 Hz, ±50 V, 3 ms squared pulses) or placebo treatment (sham stimulation 30 min/day). Forty matched healthy persons (no treatment) were included as reference. Intensity and inter-hand coherence related measures were extracted. RESULTS The T-PEMF treatment decreased the inter-hand coherence in the PD group with unilateral postural tremor. The PD group with unilateral postural tremor was less clinically affected by the disease than the PD group with bilateral postural tremor. However, no differences between T-PEMF and placebo treatment on either intensity related or coherence related measures were found when all persons with PD were included in the analyses. The peak power decreased and the tremor intensity tended to decrease in both treatment groups. CONCLUSIONS Eight weeks of T-PEMF treatment decreased inter-hand coherence in the PD group with unilateral postural tremor, while no effects of T-PEMF treatment were found for the entire PD group. The unilateral postural tremor group was less clinically affected than the bilateral postural tremor group, suggesting that early treatment initiation may be beneficial. In theory, a reduced inter-hand coherence could result from a neuronal treatment response increasing inter-hemispheric inhibition. However, this requires further studies to determine. Studies of even longer treatment periods would be of interest. TRIAL REGISTRATION ClinicalTrials.gov , NCT02125032. Registered 29 April 2014, https://clinicaltrials.gov/ct2/show/NCT02125032?term=NCT02125032&rank=1.
Collapse
Affiliation(s)
- Anne Sofie Bøgh Malling
- Department of Neurology, Odense University Hospital, University of Southern Denmark, Odense, Denmark.
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark.
| | - Bo Mohr Morberg
- Department of Neurology, Odense University Hospital, University of Southern Denmark, Odense, Denmark
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Lene Wermuth
- Department of Neurology, Odense University Hospital, University of Southern Denmark, Odense, Denmark
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Ole Gredal
- The Danish Rehabilitation Centre for Neuromuscular Diseases, Taastrup, Denmark
| | - Per Bech
- Psychiatric Research Unit, Psychiatric Centre North Zealand, University of Copenhagen, Hillerød, Denmark
| | - Bente Rona Jensen
- Department of Neurology, Odense University Hospital, University of Southern Denmark, Odense, Denmark
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| |
Collapse
|
23
|
Malling ASB, Morberg BM, Wermuth L, Gredal O, Bech P, Jensen BR. Effect of transcranial pulsed electromagnetic fields (T-PEMF) on functional rate of force development and movement speed in persons with Parkinson's disease: A randomized clinical trial. PLoS One 2018; 13:e0204478. [PMID: 30252895 PMCID: PMC6155540 DOI: 10.1371/journal.pone.0204478] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Accepted: 09/06/2018] [Indexed: 11/18/2022] Open
Abstract
Background Parkinson’s disease is caused by dopaminergic neurodegeneration resulting in motor impairments as slow movement speed and impaired balance and coordination. Pulsed electromagnetic fields are suggested to have neuroprotective effects, and could alleviate symptoms. Objective To study 1) effects of 8-week daily transcranial pulsed electromagnetic field treatment on functional rate of force development and movement speed during two motor tasks with different levels of complexity, 2) if treatment effects depend on motor performance at baseline. Methods Ninety-seven persons with Parkinson’s disease were randomized to active transcranial pulsed electromagnetic field (squared bipolar 3 ms pulses, 50 Hz) or placebo treatment with homebased treatment 30 min/day for 8 weeks. Functional rate of force development and completion time of a sit-to-stand and a dynamic postural balance task were assessed pre and post intervention. Participants were sub-grouped in high- and low-performers according to their baseline motor performance level. Repeated measure ANOVAs were used. Results Active treatment tended to improve rate of force development during chair rise more than placebo (P = 0.064). High-performers receiving active treatment improved rate of force development during chair rise more than high-performers receiving placebo treatment (P = 0.049, active/placebo: 11.9±1.1 to 12.5±1.9 BW/s ≈ 5% / 12.4±1.3 to 12.2±1.3 BW/s, no change). No other between-treatment-group or between-treatment-subgroup differences were found. Data on rate of force development of the dynamic balance task and completion times of both motor tasks improved but did not allow for between-treatment differentiation. Conclusion Treatment with transcranial pulsed electromagnetic fields was superior to placebo regarding functional rate of force development during chair rise among high-performers. Active treatment tended to increase functional rate of force development while placebo did not. Our results suggest that mildly affected persons with Parkinson’s disease have a larger potential for neural rehabilitation than more severely affected persons and indicate that early treatment initiation may be beneficial.
Collapse
Affiliation(s)
- Anne Sofie Bøgh Malling
- Department of Neurology, Odense University Hospital, University of Southern Denmark, Odense, Denmark
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark
- * E-mail: (ASBM); (BRJ)
| | - Bo Mohr Morberg
- Department of Neurology, Odense University Hospital, University of Southern Denmark, Odense, Denmark
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Lene Wermuth
- Department of Neurology, Odense University Hospital, University of Southern Denmark, Odense, Denmark
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Ole Gredal
- The Danish Rehabilitation Centre for Neuromuscular Diseases, Taastrup, Denmark
| | - Per Bech
- Psychiatric Research Unit, Psychiatric Centre North Zealand, University of Copenhagen, Hillerød, Denmark
| | - Bente Rona Jensen
- Department of Neurology, Odense University Hospital, University of Southern Denmark, Odense, Denmark
- * E-mail: (ASBM); (BRJ)
| |
Collapse
|
24
|
Jensen BR, Malling ASB, Morberg BM, Gredal O, Bech P, Wermuth L. Effects of Long-Term Treatment with T-PEMF on Forearm Muscle Activation and Motor Function in Parkinson's Disease. Case Rep Neurol 2018; 10:242-251. [PMID: 30283322 PMCID: PMC6167712 DOI: 10.1159/000492486] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Accepted: 07/24/2018] [Indexed: 11/27/2022] Open
Abstract
Bipolar pulsed electromagnetic stimulation applied to the brain (T-PEMF) is a non-pharmacological treatment which has been shown to stimulate nerve growth, attenuate nerve abnormalities, and improve microcirculation. We report on a 62-year-old, medically well-treated man with idiopathic Parkinson's disease. He was treated with T-PEMF, 30 min per day for three 8-week periods separated by two 1-week breaks. The disease made his handwriting impossible to read mainly due to small letters and lack of fluency. Forearm EMG measured during standardized conditions showed an involuntary spiky EMG pattern with regular burst activity (on his left side) at baseline. The intervention normalized the handwriting and forearm EMG. The UPDRS-motor score decreased from 25 to 17, and UPDRS-II-handwriting decreased from a pre-intervention value of 3 to 0 after the intervention. Finally, the patient reported improved fine motor function, less muscle stiffness, less muscle cramps and tingling, and less fatigue during the day in response to the T-PEMF treatment. The improved handwriting lasted for approximately 3 months after the treatment. Our results should be considered as preliminary, and large-scale, controlled studies are recommended to elucidate the therapeutic potential of long-term treatment with T-PEMF.
Collapse
Affiliation(s)
- Bente Rona Jensen
- Department of Neurology, Odense University Hospital, University of Southern Denmark, Odense, Denmark.,Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Anne Sofie Bøgh Malling
- Department of Neurology, Odense University Hospital, University of Southern Denmark, Odense, Denmark.,Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Bo Mohr Morberg
- Department of Neurology, Odense University Hospital, University of Southern Denmark, Odense, Denmark.,Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Ole Gredal
- The Danish Rehabilitation Centre for Neuromuscular Diseases, Taastrup, Denmark
| | - Per Bech
- Psychiatric Research Unit, Psychiatric Centre North Zealand, University of Copenhagen, Hillerød, Denmark
| | - Lene Wermuth
- Department of Neurology, Odense University Hospital, University of Southern Denmark, Odense, Denmark.,Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| |
Collapse
|
25
|
Song K, Im SH, Yoon YJ, Kim HM, Lee HJ, Park GS. A 60 Hz uniform electromagnetic field promotes human cell proliferation by decreasing intracellular reactive oxygen species levels. PLoS One 2018; 13:e0199753. [PMID: 30011321 PMCID: PMC6047776 DOI: 10.1371/journal.pone.0199753] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Accepted: 06/13/2018] [Indexed: 12/27/2022] Open
Abstract
Previously, we showed that exposure of human normal and cancer cells to a 6 mT, 60 Hz gradient electromagnetic field (EMF) induced genotoxicity. Here, we investigated the cellular effects of a uniform EMF. Single or repetitive exposure to a 6 mT, 60 Hz uniform EMF neither induced DNA damage nor affected cell viability in HeLa and primary IMR-90 fibroblasts. However, continuous exposure of these cells to an EMF promoted cell proliferation. Cell viability increased 24.4% for HeLa and 15.2% for IMR-90 cells after a total 168 h exposure by subculture. This increase in cell proliferation was directly correlated with EMF strength and exposure time. When further incubated without EMF, cell proliferation slowed down to that of unexposed cells, suggesting that the proliferative effect is reversible. The expression of cell cycle markers increased in cells continuously exposed to an EMF as expected, but the distribution of cells in each stage of the cell cycle did not change. Notably, intracellular reactive oxygen species levels decreased and phosphorylation of Akt and Erk1/2 increased in cells exposed to an EMF, suggesting that reduced levels of intracellular reactive oxygen species play a role in increased proliferation. These results demonstrate that EMF uniformity at an extremely low frequency (ELF) is an important factor in the cellular effects of ELF-EMF.
Collapse
Affiliation(s)
- Kiwon Song
- Department of Biochemistry, College of Life Science & Biotechnology, Yonsei University, Seoul, Korea
- * E-mail: (KS); (GSP)
| | - Sang Hyeon Im
- Department of Electrical Engineering, Pusan National University, Pusan, Korea
| | - Yeo Jun Yoon
- Department of Biochemistry, College of Life Science & Biotechnology, Yonsei University, Seoul, Korea
| | - Hui Min Kim
- Department of Electrical Engineering, Pusan National University, Pusan, Korea
| | - Hae June Lee
- Department of Electrical Engineering, Pusan National University, Pusan, Korea
| | - Gwan Soo Park
- Department of Electrical Engineering, Pusan National University, Pusan, Korea
- * E-mail: (KS); (GSP)
| |
Collapse
|
26
|
Lei Y, Su J, Xu H, Yu Q, Zhao M, Tian J. Pulsed electromagnetic fields inhibit osteoclast differentiation in RAW264.7 macrophages via suppression of the protein kinase B/mammalian target of rapamycin signaling pathway. Mol Med Rep 2018; 18:447-454. [PMID: 29749519 DOI: 10.3892/mmr.2018.8999] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2017] [Accepted: 04/17/2018] [Indexed: 11/06/2022] Open
Abstract
When bone resorption, aided by the activity of osteoclasts, exceeds bone formation induced by osteoblasts, bone metabolism loses equilibration, which results in the development of bone diseases, including osteoporosis. Pulsed electromagnetic fields (PEMFs) are known to be involved in various biological processes, including cell proliferation, differentiation and apoptosis. However, the exact mechanism of action of osteoclasts remains poorly understood. In the present study, the effects of PEMFs on osteoclast differentiation and associated signaling pathways were systematically investigated in RAW264.7 macrophages. RAW264.7 cells were induced by receptor activator of nuclear factor‑κB ligand (RANKL) to obtain osteoclasts in vitro. The results of the present study demonstrated that PEMF exposure decreased osteoclast formation, limited tartrate‑resistant acid phosphatase activity, contracted bone resorption area and inhibited osteoclastic specific gene and protein expression. Furthermore, western blot analysis indicated that PEMFs distinctly abolished the upregulation of phosphorylated‑protein kinase B (Akt), ‑mammalian target of rapamycin (mTOR) and ‑ribosome S6 protein kinase (p70S6K) induced by RANKL, which was consistent with the effects of pharmacological inhibitor perifosine and rapamycin. Therefore, the present study suggested that PEMFs reduced osteoclast formation from RAW264.7 macrophages via inhibition of the Akt/mTOR signaling pathway. These findings provided novel insight into the mechanisms through which PEMFs suppress osteoclast differentiation.
Collapse
Affiliation(s)
- Yutian Lei
- Department of Orthopaedics, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510280, P.R. China
| | - Jinyu Su
- Department of Pathophysiology, Key Laboratory for Shock and Microcirculation Research of Guangdong, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Haixia Xu
- Department of Orthopaedics, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510280, P.R. China
| | - Qiang Yu
- Department of Orthopaedics, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510280, P.R. China
| | - Ming Zhao
- Department of Pathophysiology, Key Laboratory for Shock and Microcirculation Research of Guangdong, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Jing Tian
- Department of Orthopaedics, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510280, P.R. China
| |
Collapse
|
27
|
Hu C, Li L. Preconditioning influences mesenchymal stem cell properties in vitro and in vivo. J Cell Mol Med 2018; 22:1428-1442. [PMID: 29392844 PMCID: PMC5824372 DOI: 10.1111/jcmm.13492] [Citation(s) in RCA: 296] [Impact Index Per Article: 42.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Accepted: 10/31/2017] [Indexed: 12/15/2022] Open
Abstract
Various diseases and toxic factors easily impair cellular and organic functions in mammals. Organ transplantation is used to rescue organ function, but is limited by scarce resources. Mesenchymal stem cell (MSC)-based therapy carries promising potential in regenerative medicine because of the self-renewal and multilineage potency of MSCs; however, MSCs may lose biological functions after isolation and cultivation for a long time in vitro. Moreover, after they are injected in vivo and migrate into the damaged tissues or organs, they encounter a harsh environment coupled with death signals due to the inadequate tensegrity structure between the cells and matrix. Preconditioning, genetic modification and optimization of MSC culture conditions are key strategies to improve MSC functions in vitro and in vivo, and all of these procedures will contribute to improving MSC transplantation efficacy in tissue engineering and regenerative medicine. Preconditioning with various physical, chemical and biological factors is possible to preserve the stemness of MSCs for further application in studies and clinical tests. In this review, we mainly focus on preconditioning and the corresponding mechanisms for improving MSC activities in vitro and in vivo; we provide a glimpse into the promotion of MSC-based cell therapy development for regenerative medicine. As a promising consequence, MSC transplantation can be applied for the treatment of some terminal diseases and can prolong the survival time of patients in the near future.
Collapse
Affiliation(s)
- Chenxia Hu
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious DiseasesState Key Laboratory for Diagnosis and Treatment of Infectious DiseasesSchool of MedicineFirst Affiliated HospitalZhejiang UniversityHangzhouZhejiangChina
| | - Lanjuan Li
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious DiseasesState Key Laboratory for Diagnosis and Treatment of Infectious DiseasesSchool of MedicineFirst Affiliated HospitalZhejiang UniversityHangzhouZhejiangChina
| |
Collapse
|
28
|
Peng L, Shu X, Lang C, Yu X. Cardiotrophin-1 stimulates the neural differentiation of human umbilical cord blood-derived mesenchymal stem cells and survival of differentiated cells through PI3K/Akt-dependent signaling pathways. Cytotechnology 2017; 69:933-941. [PMID: 28601931 DOI: 10.1007/s10616-017-0103-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Accepted: 04/22/2017] [Indexed: 12/31/2022] Open
Abstract
Cardiotrophin-1 (CT1) plays an important role in the differentiation, development, and survival of neural stem cells. In this study, we analyzed its effects on the stimulation of human umbilical cord blood-derived mesenchymal stem cells in terms of their potential to differentiate into neuron-like cells, their survival characteristics, and the molecular mechanisms involved. The treatment of cells with neural induction medium (NIM) and CT1 generated more cells that were neuron-like and produced stronger expression of neural-lineage markers than cells treated with NIM and without CT1. Bcl-2 and Akt phosphorylation (p-Akt) expression levels increased significantly in cells treated with both NIM and CT1. This treatment also effectively blocked cell death following neural induction and decreased Bax, Bak and cleaved-caspase 3 expression compared with cells treated with NIM without CT1. In addition, the inhibition of phosphatidylinositol 3-kinase (PI3K) abrogated p-Akt and Bcl-2 expression. Thus, PI3K/Akt contribute to CT1-stimulated neural differentiation and to the survival of differentiated cells.
Collapse
Affiliation(s)
- Longying Peng
- Department of Pediatric, First Affiliated Hospital, Zunyi Medical College, Zunyi, 563003, Guizhou Province, China
| | - Xiaomei Shu
- Department of Pediatric, First Affiliated Hospital, Zunyi Medical College, Zunyi, 563003, Guizhou Province, China.
| | - Changhui Lang
- Department of Pediatric, First Affiliated Hospital, Zunyi Medical College, Zunyi, 563003, Guizhou Province, China
| | - Xiaohua Yu
- Department of Pediatric, First Affiliated Hospital, Zunyi Medical College, Zunyi, 563003, Guizhou Province, China
| |
Collapse
|
29
|
Zhu S, Ge J, Liu Z, Liu L, Jing D, Ran M, Wang M, Huang L, Yang Y, Huang J, Luo Z. Circadian Rhythm Influences the Promoting Role of Pulsed Electromagnetic Fields on Sciatic Nerve Regeneration in Rats. Front Neurol 2017; 8:101. [PMID: 28360885 PMCID: PMC5350136 DOI: 10.3389/fneur.2017.00101] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Accepted: 02/28/2017] [Indexed: 12/21/2022] Open
Abstract
Circadian rhythm (CR) plays a critical role in the treatment of several diseases. However, the role of CR in the treatment of peripheral nerve defects has not been studied. It is also known that the pulsed electromagnetic fields (PEMF) can provide a beneficial microenvironment to quicken the process of nerve regeneration and to enhance the quality of reconstruction. In this study, we evaluate the impact of CR on the promoting effect of PEMF on peripheral nerve regeneration in rats. We used the self-made “collagen-chitosan” nerve conduits to bridge the 15-mm nerve gaps in Sprague-Dawley rats. Our results show that PEMF stimulation at daytime (DPEMF) has most effective outcome on nerve regeneration and rats with DPEMF treatment achieve quickly functional recovery after 12 weeks. These findings indicate that CR is an important factor that determines the promoting effect of PEMF on peripheral nerve regeneration. PEMF exposure in the daytime enhances the functional recovery of rats. Our study provides a helpful guideline for the effective use of PEMF mediations experimentally and clinically.
Collapse
Affiliation(s)
- Shu Zhu
- Institute of Orthopaedics, Xijing Hospital, The Fourth Military Medical University , Xi'an , China
| | - Jun Ge
- Department of Orthopaedics, 323rd Hospital of PLA, Xi'an, China; Department of Anatomy, The Fourth Military Medical University, Xi'an, China
| | - Zhongyang Liu
- Institute of Orthopaedics, Xijing Hospital, The Fourth Military Medical University , Xi'an , China
| | - Liang Liu
- Department of Orthopaedics, 161st Hospital of PLA , Wuhan , China
| | - Da Jing
- Faculty of Biomedical Engineering, Fourth Military Medical University , Xi'an , China
| | - Mingzi Ran
- Department of Anesthesiology, Xijing Hospital, The Fourth Military Medical University , Xi'an , China
| | - Meng Wang
- General Political Department Hospital of PLA , Beijing , China
| | - Liangliang Huang
- Institute of Orthopaedics, Xijing Hospital, The Fourth Military Medical University , Xi'an , China
| | - Yafeng Yang
- Institute of Orthopaedics, Xijing Hospital, The Fourth Military Medical University , Xi'an , China
| | - Jinghui Huang
- Institute of Orthopaedics, Xijing Hospital, The Fourth Military Medical University , Xi'an , China
| | - Zhuojing Luo
- Institute of Orthopaedics, Xijing Hospital, The Fourth Military Medical University , Xi'an , China
| |
Collapse
|
30
|
Kim YM, Cho SE, Seo YK. The activation of melanogenesis by p-CREB and MITF signaling with extremely low-frequency electromagnetic fields on B16F10 melanoma. Life Sci 2016; 162:25-32. [PMID: 27543340 DOI: 10.1016/j.lfs.2016.08.015] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2016] [Revised: 07/28/2016] [Accepted: 08/14/2016] [Indexed: 10/21/2022]
Abstract
Melanin in the skin determines the skin color, and decreased melanin causes many hypopigmentation disorders and increased damage to skin by ultraviolet B (UVB) light irradiation. Here, we stimulate melanogenesis in B16F10 melanoma cells by using specific frequencies of ELF-EMFs. In this study, we focus on the melanogenesis of EMF-ELFs and find that 60-75Hz ELF-EMFs upregulate melanin synthesis by stimulated expression of tyrosinase and TRP-1 through inhibition of phosphorylation ERK, activation of CREB, and MITF up-regulation in B16F10 melanoma cells. The results show that 60-75Hz ELF-EMFs significantly increase secreted melanin, cellular melanin content, and tyrosinase activity, and the cell mitochondria activity, cell viability, and cell membrane condition are unchanged. Furthermore, the protein expression level of MITF and p-CREB signaling pathway are significantly increased. Moreover, 60Hz ELF-EMFs reduce the phosphorylate of ERK in B16F10 melanoma cells. These findings indicate that stimulation of melanogenesis by using ELF-EMFs has therapeutic potential for treating hypopigmentation disorders such as vitiligo.
Collapse
Affiliation(s)
- Yu-Mi Kim
- Department of Medical Biotechnology (BK21 Plus team), Dongguk University, Seoul 100-715, Republic of Korea
| | - Sang-Eun Cho
- Department of Medical Biotechnology (BK21 Plus team), Dongguk University, Seoul 100-715, Republic of Korea
| | - Young-Kwon Seo
- Department of Medical Biotechnology (BK21 Plus team), Dongguk University, Seoul 100-715, Republic of Korea.
| |
Collapse
|