1
|
Kaadan A, Salati S, Cadossi R, Aaron R. Regulation of Inflammatory Responses by Pulsed Electromagnetic Fields. Bioengineering (Basel) 2025; 12:474. [PMID: 40428093 DOI: 10.3390/bioengineering12050474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2025] [Revised: 04/21/2025] [Accepted: 04/23/2025] [Indexed: 05/29/2025] Open
Abstract
Pulsed Electromagnetic Field (PEMF) therapy has been shown to have substantial suppressive effects on inflammation and is a promising treatment for the modulation of inflammation. Several in vitro and in vivo studies have shown that PEMFs profoundly suppress inflammatory pathways, such as the NF-κB and MAPK signaling pathways, by lowering cytokine levels and improving extracellular matrix synthesis. This review describes studies, ranging from in vitro to clinical, that investigate the lesser-known roles of PEMF in the modulation of inflammation in soft tissue wound, cartilage, and joint healing, alongside angiogenesis. Mechanistically, PEMFs act via adenosine receptors, specifically A2A, which play a key role in inflammation modulation and tissue repair. In some clinical trials, PEMF has yielded short-term symptom relief and functional improvements in early-stage osteoarthritis patients, arthroscopy patients, and anterior cruciate ligament reconstruction patients.
Collapse
Affiliation(s)
- Amr Kaadan
- Department of Orthopaedic Surgery, Warren Alpert Medical School of Brown University, Providence, RI 02903, USA
| | | | | | - Roy Aaron
- Department of Orthopaedic Surgery, Warren Alpert Medical School of Brown University, Providence, RI 02903, USA
| |
Collapse
|
2
|
Zhi W, Tang J, Zhang M, Zou Y, Qiao S, Ma L, Dong J, Yao B, Zhao X, Yang Z, Lin Z, Hu X, Wang L. Mechanistic insights into microwave radiation induced cognitive impairments: The role of m 6A epigenetic modifications and HNRNPA2B1 in TrkB regulation. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2025; 292:117907. [PMID: 39999628 DOI: 10.1016/j.ecoenv.2025.117907] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 02/14/2025] [Accepted: 02/14/2025] [Indexed: 02/27/2025]
Abstract
Microwave radiation, a prevalent environmental stressor, significantly impacts human health. Based on previous studies, we hypothesize that microwave-induced cognitive impairments and vulnerability in the hippocampal dentate gyrus (DG) region are due to abnormal synaptic plasticity regulated by both newborn and mature neurons derived from neural stem cells (NSCs). Epigenetics links external factors to organisms, offers insights into the health effects of environmental influences. To explore the molecular mechanisms underlying the effects of microwave radiation on neuronal synaptic plasticity from the perspective of mRNA N6-methyladenosine (m6A) modification. We first assessed the impact of microwave radiation on cognitive memory abilities in rats through behavioral tests. Immunofluorescence staining were applied to clarify the influence of microwave radiation on both neurons and NSCs. Molecular mechanisms were investigated by ELISA, q-PCR, Western blot, MeRIP-seq, and RNA pull-down experiments. The microwave radiated rat model exhibiting learning and memory deficits. Impaired synaptic plasticity in mature hippocampal neurons alongside hindered NSCs proliferation and development were observed. Using our established non-contact co-culture model, we replicated the in vivo adverse effects of microwave radiation. Down-regulated HNRNPA2B1 leads to reduced binding of TrkB m6A and promoted TrkB degradation. This feedback loop results in low BDNF expression, ultimately causing cognitive impairments. Our study emphasizes the neurotoxicity of microwave radiation and identifies TrkB m6A modification as a potential target for protecting against cognitive damage induced by electromagnetic radiation.
Collapse
Affiliation(s)
- Weijia Zhi
- Beijing Institute of Radiation Medicine, Beijing, China.
| | - Jiale Tang
- Changsha Hospital of Traditional Chinese Medicine (Changsha Eighth Hospital), Changsha, China.
| | - Mingzhao Zhang
- Beijing Institute of Radiation Medicine, Beijing, China.
| | - Yong Zou
- Beijing Institute of Radiation Medicine, Beijing, China.
| | - Simo Qiao
- Beijing Institute of Pharmacology and Toxicology, Beijing, China.
| | - Lizhen Ma
- Beijing Institute of Radiation Medicine, Beijing, China.
| | - Ji Dong
- Beijing Institute of Radiation Medicine, Beijing, China.
| | - Binwei Yao
- Beijing Institute of Radiation Medicine, Beijing, China.
| | - Xuelong Zhao
- Beijing Institute of Radiation Medicine, Beijing, China.
| | - Zhenqi Yang
- Beijing Institute of Radiation Medicine, Beijing, China.
| | - Zhongwu Lin
- Beijing Institute of Radiation Medicine, Beijing, China; National Innovation Institute of Defense Technology, Beijing, China.
| | - Xiangjun Hu
- Beijing Institute of Radiation Medicine, Beijing, China.
| | - Lifeng Wang
- Beijing Institute of Radiation Medicine, Beijing, China.
| |
Collapse
|
3
|
Isaković J, Chin BD, Oberwinter M, Rance HK. From lab coats to clinical trials: Evolution and application of electromagnetic fields for ischemic stroke rehabilitation and monitoring. Brain Res 2025; 1850:149391. [PMID: 39662791 DOI: 10.1016/j.brainres.2024.149391] [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: 08/15/2024] [Revised: 12/05/2024] [Accepted: 12/06/2024] [Indexed: 12/13/2024]
Abstract
Stroke is a neurovascular disorder which stands as one of the leading causes of death and disability worldwide, resulting in motor and cognitive impairment. Although the treatment approach depends on the time elapsed, the type of stroke and the availability of care centers, common interventions include thrombectomy or the administration of a tissue plasminogen activator (tPA). While these methods restore blood flow, they fall short in helping patients regain lost function. With that, recent years have seen a rise in novel methods, one of which is the use of electromagnetic fields (EMFs). Due to their ability to impact the charges in their vicinity, thereby altering the immune response and cell signaling, EMFs became suitable candidates for stroke rehabilitation. Based on their characteristics, therapeutic EMFs can be categorized into transcranial magnetic stimulation (TMS), transcranial direct current stimulation (tDCS), pulsed (PEMFs) and low frequency (LF-EMFs) electromagnetic fields, among others. In addition to treatment, EMFs are being explored for stroke monitoring, utilizing external EMFs for imaging or recording innate EMFs linked to neural activity. Drawing from research on the effects of EMFs, this review aims to provide a comprehensive overview of the physical principles and molecular mechanisms underlying the action of EMFs, along with a discussion of their application in preclinical studies and clinical trials. Finally, this paper not only addresses the importance of treatment availability and potential side-effects, but also delves into the technical and ethical challenges associated with the use of EMFs, while exploring their prospects and future opportunities.
Collapse
Affiliation(s)
- Jasmina Isaković
- School of Medicine, European University Cyprus - Frankfurt Branch, 60488 Frankfurt am Main, Germany.
| | - Benjamin Daniel Chin
- School of Medicine, European University Cyprus - Frankfurt Branch, 60488 Frankfurt am Main, Germany
| | - Moritz Oberwinter
- School of Medicine, European University Cyprus - Frankfurt Branch, 60488 Frankfurt am Main, Germany
| | - Hannah Katarina Rance
- School of Medicine, European University Cyprus - Frankfurt Branch, 60488 Frankfurt am Main, Germany
| |
Collapse
|
4
|
Merighi S, Nigro M, Travagli A, Fernandez M, Vincenzi F, Varani K, Pasquini S, Borea PA, Salati S, Cadossi R, Gessi S. Effect of Low-Frequency, Low-Energy Pulsed Electromagnetic Fields in Neuronal and Microglial Cells Injured with Amyloid-Beta. Int J Mol Sci 2024; 25:12847. [PMID: 39684558 DOI: 10.3390/ijms252312847] [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: 10/09/2024] [Revised: 11/26/2024] [Accepted: 11/27/2024] [Indexed: 12/18/2024] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative pathology covering about 70% of all cases of dementia. It is associated with neuroinflammation and neuronal cell death, which are involved in disease progression. There is a lack of effective therapies, and halting this process represents a therapeutic challenge. Data in the literature suggest several neuroprotective effects of low-frequency, low-energy pulsed electromagnetic fields (PEMFs) on biological systems, and clinical studies report that PEMF stimulation is safe and well tolerated. The aim of this work is to investigate the effects of PEMF exposure on oxidative stress and cell death in in vitro-injured cellular models of neurons and microglia. SH-SY5Y cells were stimulated by hydrogen peroxide (H2O2) or amyloid-β (Aβ) peptide, and N9 microglial cells were activated with lipopolysaccharide (LPS) or Aβ peptide. Reactive oxygen production, mitochondrial integrity, and cell death modulation were investigated through 2',7'-dichlorodihydrofluorescein diacetate (H2DCFDA) and 5,5',6,6'-tetrachloro-1,1',3,3'-tetraethylbenzimidazolocarbo-cyanine iodide (JC-1) biochemical assays, fluorescence, and MTS experiments. Cells were exposed to PEMFs producing a pulsed signal with the following parameters: pulse duration of 1.3 ms and frequency of 75 Hz. The outcomes demonstrated that PEMFs defended SH-SY5Y cells against Aβ peptide- or H2O2-induced oxidative stress, mitochondrial damage, and cell death. Furthermore, in microglia activated by LPS or Aβ peptide, they reverted the reduction in mitochondrial potential, oxidative damage, and cell death. Overall, these findings imply that PEMFs influence the redox state of the cells by significantly boosting antioxidant levels in both injured microglia and neuronal in vitro cells mimicking in vitro AD.
Collapse
Affiliation(s)
- Stefania Merighi
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy
| | - Manuela Nigro
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy
| | - Alessia Travagli
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy
| | - Mercedes Fernandez
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy
| | - Fabrizio Vincenzi
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy
| | - Katia Varani
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy
| | - Silvia Pasquini
- Department of Chemical, Pharmaceutical and Agricultural Science, University of Ferrara, 44121 Ferrara, Italy
| | | | - Simona Salati
- Igea Clinical Biophysics, Medical Division, 41012 Carpi, Italy
| | - Ruggero Cadossi
- Igea Clinical Biophysics, Medical Division, 41012 Carpi, Italy
| | - Stefania Gessi
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy
| |
Collapse
|
5
|
Siwak M, Piotrzkowska D, Skrzypek M, Majsterek I. Effects of PEMF and LIPUS Therapy on the Expression of Genes Related to Peripheral Nerve Regeneration in Schwann Cells. Int J Mol Sci 2024; 25:12791. [PMID: 39684499 DOI: 10.3390/ijms252312791] [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: 11/02/2024] [Revised: 11/26/2024] [Accepted: 11/26/2024] [Indexed: 12/18/2024] Open
Abstract
Peripheral nerve regeneration remains a major challenge in neuroscience, despite advancements in understanding its mechanisms. Current treatments, including nerve transplantation and drug therapies, face limitations such as invasiveness and incomplete recovery of nerve function. Physical therapies, like pulsed electromagnetic fields (PEMF) and low-intensity ultrasound (LIPUS), are gaining attention for their potential to enhance regeneration. This study analyzes the effects of PEMF and LIPUS on gene expression in human primary Schwann cells, which are crucial for nerve myelination and repair. Key genes involved in neurotrophin signaling (NGF, BDNF), inflammation (IL-1β, IL-6, IL-10, TNF-α, TGF-β), and regeneration (CRYAB, CSPG, Ki67) were assessed. The results of this study reveal that combined PEMF and LIPUS therapies promote Schwann cell proliferation, reduce inflammation, and improve the regenerative environment, offering potential for optimizing these therapies for clinical use in regenerative medicine.
Collapse
Affiliation(s)
- Mateusz Siwak
- Department of Clinical Chemistry and Biochemistry, Medical University of Lodz, 92-215 Lodz, Poland
| | - Danuta Piotrzkowska
- Department of Clinical Chemistry and Biochemistry, Medical University of Lodz, 92-215 Lodz, Poland
| | - Maciej Skrzypek
- Department of Clinical Chemistry and Biochemistry, Medical University of Lodz, 92-215 Lodz, Poland
| | - Ireneusz Majsterek
- Department of Clinical Chemistry and Biochemistry, Medical University of Lodz, 92-215 Lodz, Poland
| |
Collapse
|
6
|
Marchewka R, Trzmiel T, Hojan K. The Effect of Extremely Low-Frequency Magnetic Field on Stroke Patients: A Systematic Review. Brain Sci 2024; 14:430. [PMID: 38790409 PMCID: PMC11119128 DOI: 10.3390/brainsci14050430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 04/23/2024] [Accepted: 04/24/2024] [Indexed: 05/26/2024] Open
Abstract
BACKGROUND The aim of this study was to review the current state of scientific evidence on the effect of extremely low-frequency magnetic fields stimulation (ELF-MFs) on stroke patients. METHODS A systematic review of PubMed, ScienceDirect, PeDro and Embase databases was conducted. Only articles published in English, involving adult participants and focusing on individuals who had experienced a stroke, specifically examining the impact of ELF-MFs on post-stroke patients and had well-defined criteria for inclusion and exclusion of participants, were included. The methodological quality of the included studies was assessed using the Quality Assessment Tool for Quantitative Studies (QATQS). RESULTS A total of 71 studies were identified through database and reference lists' search, from which 9 were included in the final synthesis. All included studies showed a beneficial effect of ELF-MFs on stroke patients, however seven of the included studies were carried by the same research group. Improvements were observed in domains such as oxidative stress, inflammation, ischemic lesion size, functional status, depressive symptoms and cognitive abilities. CONCLUSIONS The available literature suggests a beneficial effect of ELF-MFs on post-stroke patients; however, the current data are too limited to broadly recommend the use of this method. Further research with improved methodological quality is necessary.
Collapse
Affiliation(s)
- Renata Marchewka
- Neurorehabilitation Ward, Greater Poland Provincial Hospital, 60-480 Poznan, Poland; (R.M.); (K.H.)
| | - Tomasz Trzmiel
- Department of Occupational Therapy, Poznan University of Medical Sciences, 60-781 Poznan, Poland
| | - Katarzyna Hojan
- Neurorehabilitation Ward, Greater Poland Provincial Hospital, 60-480 Poznan, Poland; (R.M.); (K.H.)
- Department of Occupational Therapy, Poznan University of Medical Sciences, 60-781 Poznan, Poland
- Department of Rehabilitation, Greater Poland Cancer Centre, 61-866 Poznan, Poland
| |
Collapse
|
7
|
Isaković J, Slatković F, Jagečić D, Petrović DJ, Mitrečić D. Pulsating Extremely Low-Frequency Electromagnetic Fields Influence Differentiation of Mouse Neural Stem Cells towards Astrocyte-like Phenotypes: In Vitro Pilot Study. Int J Mol Sci 2024; 25:4038. [PMID: 38612847 PMCID: PMC11012476 DOI: 10.3390/ijms25074038] [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: 02/29/2024] [Revised: 03/26/2024] [Accepted: 04/03/2024] [Indexed: 04/14/2024] Open
Abstract
Even though electromagnetic fields have been reported to assist endogenous neurogenesis, little is known about the exact mechanisms of their action. In this pilot study, we investigated the effects of pulsating extremely low-frequency electromagnetic fields on neural stem cell differentiation towards specific phenotypes, such as neurons and astrocytes. Neural stem cells isolated from the telencephalic wall of B6(Cg)-Tyrc-2J/J mouse embryos (E14.5) were randomly divided into three experimental groups and three controls. Electromagnetic field application setup included a solenoid placed within an incubator. Each of the experimental groups was exposed to 50Hz ELF-EMFs of varied strengths for 1 h. The expression of each marker (NES, GFAP, β-3 tubulin) was then assessed by immunocytochemistry. The application of high-strength ELF-EMF significantly increased and low-strength ELF-EMF decreased the expression of GFAP. A similar pattern was observed for β-3 tubulin, with high-strength ELF-EMFs significantly increasing the immunoreactivity of β-3 tubulin and medium- and low-strength ELF-EMFs decreasing it. Changes in NES expression were observed for medium-strength ELF-EMFs, with a demonstrated significant upregulation. This suggests that, even though ELF-EMFs appear to inhibit or promote the differentiation of neural stem cells into neurons or astrocytes, this effect highly depends on the strength and frequency of the fields as well as the duration of their application. While numerous studies have demonstrated the capacity of EMFs to guide the differentiation of NSCs into neuron-like cells or β-3 tubulin+ neurons, this is the first study to suggest that ELF-EMFs may also steer NSC differentiation towards astrocyte-like phenotypes.
Collapse
Affiliation(s)
| | - Filip Slatković
- Omnion Research International d.o.o., 10000 Zagreb, Croatia;
| | - Denis Jagečić
- Laboratory for Stem Cells, Croatian Institute for Brain Research, University of Zagreb School of Medicine, 10000 Zagreb, Croatia
- Department of Histology and Embryology, University of Zagreb School of Medicine, 10000 Zagreb, Croatia
| | - Dražen Juraj Petrović
- Laboratory for Stem Cells, Croatian Institute for Brain Research, University of Zagreb School of Medicine, 10000 Zagreb, Croatia
- Department of Histology and Embryology, University of Zagreb School of Medicine, 10000 Zagreb, Croatia
- Genos d.o.o., Laboratory for Glycobiology, 10000 Zagreb, Croatia
| | - Dinko Mitrečić
- Laboratory for Stem Cells, Croatian Institute for Brain Research, University of Zagreb School of Medicine, 10000 Zagreb, Croatia
- Department of Histology and Embryology, University of Zagreb School of Medicine, 10000 Zagreb, Croatia
| |
Collapse
|
8
|
Amirinejad M, Eftekhar-Vaghefi SH, Nematollahi Mahani SN, Salari M, Yahyapour R, Ahmadi-Zeidabadi M. Exposure to Low-Frequency Radiation Changes the Expression of Nestin, VEGF, BCRP and Apoptosis Markers During Glioma Treatment Strategy: An In Vitro Study. Curr Radiopharm 2024; 17:55-67. [PMID: 38817005 DOI: 10.2174/0118744710258350230921065159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 08/20/2023] [Accepted: 08/28/2023] [Indexed: 06/01/2024]
Abstract
BACKGROUND Exposure to physical contamination during chemotherapy, including non-ionizing electromagnetic fields, raises concerns about the widespread sources of exposure to this type of radiation. Glioblastoma multiforme (GBM) is an aggressive central nervous system tumor that is hard to treat due to resistance to drugs such as temozolomide (TMZ). OBJECTIVE Electromagnetic fields (EMF) and haloperidol (HLP) may have anticancer effects. In this study, we investigated the effects of TMZ, HLP, and EMF on GBM cell lines and analyzed the association between non-ionizing radiation and the risk of change in drug performance. METHODS Cell viability and reactive oxygen species (ROS) generation were measured by MTT and NBT assay, respectively. Then, the expression levels of breast cancer-resistant protein (BCRP), Bax, Bcl2, Nestin, vascular endothelial growth factor (VEGF) genes, and P53, Bax, and Bcl2 Proteins were evaluated by real-time PCR and western blot. RESULTS Co-treatment of GBM cells by HLP and TMZ enhanced apoptosis in T-98G and A172 cells by increasing the expression of P53 and Bax and decreasing Bcl-2. Interestingly, exposure of GBM cells to EMF decreased apoptosis in the TMZ+HLP group. CONCLUSION In conclusion, EMF reduced the synergistic effect of TMZ and HLP. This hypothesis that patients who are treated for brain tumors and suffer from depression should not be exposed to EMF is proposed in the present study. There appears to be an urgent need to reconsider exposure limits for low-frequency magnetic fields, based on experimental and epidemiological research, the relationship between exposure to non-ionizing radiation and adverse human health effects.
Collapse
Affiliation(s)
- Maryam Amirinejad
- Department of Anatomy, Afzalipour School of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Seyed Hassan Eftekhar-Vaghefi
- Department of Anatomy, Afzalipour School of Medicine, Kerman University of Medical Sciences, Kerman, Iran
- Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | | | | | - Rasoul Yahyapour
- School of Medicine, Jiroft University of Medical Sciences, Jiroft, Iran
| | - Meysam Ahmadi-Zeidabadi
- Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| |
Collapse
|
9
|
Chacko TP, Toole JT, Morris MC, Page J, Forsten RD, Barrett JP, Reinhard MJ, Brewster RC, Costanzo ME, Broderick G. A regulatory pathway model of neuropsychological disruption in Havana syndrome. Front Psychiatry 2023; 14:1180929. [PMID: 37965360 PMCID: PMC10642174 DOI: 10.3389/fpsyt.2023.1180929] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Accepted: 09/29/2023] [Indexed: 11/16/2023] Open
Abstract
Introduction In 2016 diplomatic personnel serving in Havana, Cuba, began reporting audible sensory phenomena paired with onset of complex and persistent neurological symptoms consistent with brain injury. The etiology of these Anomalous Health Incidents (AHI) and subsequent symptoms remains unknown. This report investigates putative exposure-symptom pathology by assembling a network model of published bio-behavioral pathways and assessing how dysregulation of such pathways might explain loss of function in these subjects using data available in the published literature. Given similarities in presentation with mild traumatic brain injury (mTBI), we used the latter as a clinically relevant means of evaluating if the neuropsychological profiles observed in Havana Syndrome Havana Syndrome might be explained at least in part by a dysregulation of neurotransmission, neuro-inflammation, or both. Method Automated text-mining of >9,000 publications produced a network consisting of 273 documented regulatory interactions linking 29 neuro-chemical markers with 9 neuropsychological constructs from the Brief Mood Survey, PTSD Checklist, and the Frontal Systems Behavior Scale. Analysis of information flow through this network produced a set of regulatory rules reconciling to within a 6% departure known mechanistic pathways with neuropsychological profiles in N = 6 subjects. Results Predicted expression of neuro-chemical markers that jointly satisfy documented pathways and observed symptom profiles display characteristically elevated IL-1B, IL-10, NGF, and norepinephrine levels in the context of depressed BDNF, GDNF, IGF1, and glutamate expression (FDR < 5%). Elevations in CRH and IL-6 were also predicted unanimously across all subjects. Furthermore, simulations of neurological regulatory dynamics reveal subjects do not appear to be "locked in" persistent illness but rather appear to be engaged in a slow recovery trajectory. Discussion This computational analysis of measured neuropsychological symptoms in Havana-based diplomats proposes that these AHI symptoms may be supported in part by disruption of known neuroimmune and neurotransmission regulatory mechanisms also associated with mTBI.
Collapse
Affiliation(s)
- Thomas P. Chacko
- Center for Clinical Systems Biology, Rochester General Hospital, Rochester, NY, United States
| | - J. Tory Toole
- Center for Clinical Systems Biology, Rochester General Hospital, Rochester, NY, United States
| | - Matthew C. Morris
- Center for Clinical Systems Biology, Rochester General Hospital, Rochester, NY, United States
| | - Jeffrey Page
- Center for Clinical Systems Biology, Rochester General Hospital, Rochester, NY, United States
| | - Robert D. Forsten
- War Related Illness and Injury Study Center (WRIISC), Department of Veterans Affairs, Washington, DC, United States
| | - John P. Barrett
- War Related Illness and Injury Study Center (WRIISC), Department of Veterans Affairs, Washington, DC, United States
- Department of Preventive Medicine and Biostatistics, Uniformed Services University, Bethesda, MD, United States
| | - Matthew J. Reinhard
- War Related Illness and Injury Study Center (WRIISC), Department of Veterans Affairs, Washington, DC, United States
- Complex Exposures Threats Center, Department of Veterans Affairs, Washington, DC, United States
| | - Ryan C. Brewster
- War Related Illness and Injury Study Center (WRIISC), Department of Veterans Affairs, Washington, DC, United States
| | - Michelle E. Costanzo
- War Related Illness and Injury Study Center (WRIISC), Department of Veterans Affairs, Washington, DC, United States
- Complex Exposures Threats Center, Department of Veterans Affairs, Washington, DC, United States
- Department of Medicine, Uniformed Services University, Bethesda, MD, United States
| | - Gordon Broderick
- Center for Clinical Systems Biology, Rochester General Hospital, Rochester, NY, United States
- Complex Exposures Threats Center, Department of Veterans Affairs, Washington, DC, United States
| |
Collapse
|
10
|
Jashire Nezhad N, Safari A, Namavar MR, Nami M, Karimi-Haghighi S, Pandamooz S, Dianatpour M, Azarpira N, Khodabandeh Z, Zare S, Hooshmandi E, Bayat M, Owjfard M, Zafarmand SS, Fadakar N, Jaberi AR, Salehi MS, Borhani-Haghighi A. Short-term beneficial effects of human dental pulp stem cells and their secretome in a rat model of mild ischemic stroke. J Stroke Cerebrovasc Dis 2023; 32:107202. [PMID: 37354874 DOI: 10.1016/j.jstrokecerebrovasdis.2023.107202] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Revised: 05/01/2023] [Accepted: 05/29/2023] [Indexed: 06/26/2023] Open
Abstract
Although cell therapy has been applied in regenerative medicine for decades, recent years have seen greatly increased attention being given to the use of stem cell-based derivatives such as cell-free secretome. Dental pulp stem cells (DPSCs) are widely available, easily accessible, and have high neuroprotective and angiogenic properties. In addition, DPSC-derived secretome contains a rich mixture of trophic factors. The current investigation evaluated the short-term therapeutic effects of human DPSCs and their secretome in a rat model of mild ischemic stroke. Mild ischemic stroke was induced by 30 min middle cerebral artery occlusion, and hDPSCs or their secretome was administered intra-arterially and intranasally. Neurological function, infarct size, spatial working memory, and relative expression of seven target genes in two categories of neurotrophic and angiogenic factors were assessed three days after stroke. In the short-term, all treatments reduced the severity of neurological and histological deficits caused by ischemic stroke. Moreover, transient middle cerebral artery occlusion led to the striatal and cortical over-expression of BDNF, NT-3, and angiogenin, while NGF and VEGF expression was reduced. Almost all interventions were able to modulate the expression of target genes after stroke. The obtained data revealed that single intra-arterial administration of hDPSCs or their secretome, single intranasal transplantation of hDPSCs, or repeated intranasal administration of hDPSC-derived secretome was able to ameliorate the devastating effects of a mild stroke, at least in the short-term.
Collapse
Affiliation(s)
- Nahid Jashire Nezhad
- Department of Neuroscience, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Anahid Safari
- Stem Cells Technology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohammad Reza Namavar
- Histomorphometry & Stereology Research Centre, Shiraz University of Medical Sciences, Shiraz, Iran; Clinical Neurology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohammad Nami
- Department of Neuroscience, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | - Sareh Pandamooz
- Stem Cells Technology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mehdi Dianatpour
- Stem Cells Technology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Negar Azarpira
- Transplant Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Zahra Khodabandeh
- Stem Cells Technology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Shahrokh Zare
- Stem Cells Technology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Etrat Hooshmandi
- Clinical Neurology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mahnaz Bayat
- Clinical Neurology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Maryam Owjfard
- Clinical Neurology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | - Nima Fadakar
- Clinical Neurology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Abbas Rahimi Jaberi
- Clinical Neurology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohammad Saied Salehi
- Clinical Neurology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
| | | |
Collapse
|
11
|
Purinergic signaling: a potential therapeutic target for ischemic stroke. Purinergic Signal 2023; 19:173-183. [PMID: 36370253 PMCID: PMC9984595 DOI: 10.1007/s11302-022-09905-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Accepted: 10/30/2022] [Indexed: 11/15/2022] Open
Abstract
Pathogenesis of ischemic stroke is mainly characterized by thrombosis and neuroinflammation. Purinergic signaling pathway constitutes adenosine triphosphate (ATP), adenosine diphosphate (ADP), adenosine monophosphate (AMP), and adenosine (ADO). ATP is hydrolyzed to ADP and then to AMP by extracellular nucleotidase CD39; AMP is subsequently converted to adenosine by CD73. All these nucleotides and nucleosides act on purinergic receptors protecting against thrombosis and inhibit inflammation. In addition, many physical methods have been found to play a neuroprotective role through purinergic signaling. This review mainly introduces the role and potential mechanism of purinergic signalings in the treatment of ischemic stroke, so as to provide reference for seeking new treatment methods for stroke.
Collapse
|
12
|
Non-invasive brain stimulation as therapeutic approach for ischemic stroke: Insights into the (sub)cellular mechanisms. Pharmacol Ther 2022; 235:108160. [PMID: 35183592 DOI: 10.1016/j.pharmthera.2022.108160] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 02/11/2022] [Accepted: 02/14/2022] [Indexed: 01/12/2023]
Abstract
Although spontaneous recovery can occur following ischemic stroke due to endogenous neuronal reorganization and neuroplastic events, the degree of functional improvement is highly variable, causing many patients to remain permanently impaired. In the last decades, non-invasive brain stimulation (NIBS) techniques have emerged as potential add-on interventions to the standard neurorehabilitation programs to improve post-stroke recovery. Due to their ability to modulate cortical excitability and to induce neuroreparative processes in the brain, multiple studies have assessed the safety, efficacy and (sub)cellular mechanisms of NIBS following ischemic stroke. In this review, an overview will be provided of the different NIBS techniques that are currently being investigated in (pre)clinical stroke studies. The NIBS therapies that will be discussed include transcranial magnetic stimulation, transcranial direct current stimulation and extremely low frequency electromagnetic stimulation. First, an overview will be given of the cellular mechanisms induced by NIBS that are associated with enhanced stroke outcome in preclinical models. Furthermore, the current knowledge on safety and efficacy of these NIBS techniques in stroke patients will be reviewed.
Collapse
|
13
|
Kemps H, Dessy C, Dumas L, Sonveaux P, Alders L, Van Broeckhoven J, Font LP, Lambrichts S, Foulquier S, Hendrix S, Brône B, Lemmens R, Bronckaers A. Extremely low frequency electromagnetic stimulation reduces ischemic stroke volume by improving cerebral collateral blood flow. J Cereb Blood Flow Metab 2022; 42:979-996. [PMID: 35209740 PMCID: PMC9125494 DOI: 10.1177/0271678x221084410] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Extremely low frequency electromagnetic stimulation (ELF-EMS) has been considered as a neuroprotective therapy for ischemic stroke based on its capacity to induce nitric oxide (NO) signaling. Here, we examined whether ELF-EMS reduces ischemic stroke volume by stimulating cerebral collateral perfusion. Moreover, the pathway responsible for ELF-EMS-induced NO production was investigated. ELF-EMS diminished infarct growth following experimental stroke in collateral-rich C57BL/6 mice, but not in collateral-scarce BALB/c mice, suggesting that decreased lesion sizes after ELF-EMS results from improved collateral blood flow. In vitro analysis demonstrated that ELF-EMS increased endothelial NO levels by stimulating the Akt-/eNOS pathway. Furthermore, ELF-EMS augmented perfusion in the hind limb of healthy mice, which was mediated by enhanced Akt-/eNOS signaling. In healthy C57BL/6 mouse brains, ELF-EMS treatment increased cerebral blood flow in a NOS-dependent manner, whereas no improvement in cerebrovascular perfusion was observed in collateral-sparse BALB/c mice. In addition, ELF-EMS enhanced cerebral blood flow in both the contra- and ipsilateral hemispheres of C57BL/6 mice subjected to experimental ischemic stroke. In conclusion, we showed that ELF-EMS enhances (cerebro)vascular perfusion by stimulating NO production, indicating that ELF-EMS could be an attractive therapeutic strategy for acute ischemic stroke by improving cerebral collateral blood flow.
Collapse
Affiliation(s)
- Hannelore Kemps
- Biomedical Research Institute (BIOMED), Hasselt University (UHasselt), Diepenbeek, Belgium
| | - Chantal Dessy
- Pole of Pharmacology and Therapeutics, Institut de Recherche Expérimentale et Clinique (IREC), Université catholique de Louvain (UCLouvain), Brussels, Belgium
| | - Laurent Dumas
- Pole of Pharmacology and Therapeutics, Institut de Recherche Expérimentale et Clinique (IREC), Université catholique de Louvain (UCLouvain), Brussels, Belgium
| | - Pierre Sonveaux
- Pole of Pharmacology and Therapeutics, Institut de Recherche Expérimentale et Clinique (IREC), Université catholique de Louvain (UCLouvain), Brussels, Belgium
| | - Lotte Alders
- Biomedical Research Institute (BIOMED), Hasselt University (UHasselt), Diepenbeek, Belgium
| | - Jana Van Broeckhoven
- Biomedical Research Institute (BIOMED), Hasselt University (UHasselt), Diepenbeek, Belgium
| | - Lena Perez Font
- Centro Nacional de Electromagnetismo Aplicado (CNEA), Universidad de Oriente, Santiago de Cuba, Cuba
| | - Sara Lambrichts
- Department of Pharmacology and Toxicology, School for Mental Health and Neuroscience, Maastricht University Medical Center+, Maastricht, The Netherlands
| | - Sébastien Foulquier
- Department of Pharmacology and Toxicology, School for Mental Health and Neuroscience, Maastricht University Medical Center+, Maastricht, The Netherlands.,CARIM, School for Cardiovascular Diseases, Maastricht University, Maastricht, The Netherlands
| | - Sven Hendrix
- Biomedical Research Institute (BIOMED), Hasselt University (UHasselt), Diepenbeek, Belgium.,Medical School Hamburg, Hamburg, Germany
| | - Bert Brône
- Biomedical Research Institute (BIOMED), Hasselt University (UHasselt), Diepenbeek, Belgium
| | - Robin Lemmens
- KU Leuven, - University of Leuven, Department of Neurosciences, Experimental Neurology, Leuven, Belgium.,VIB, Center for Brain & Disease Research, Laboratory of Neurobiology, Leuven, Belgium.,Department of Neurology, University Hospitals Leuven, Leuven, Belgium
| | - Annelies Bronckaers
- Biomedical Research Institute (BIOMED), Hasselt University (UHasselt), Diepenbeek, Belgium
| |
Collapse
|
14
|
Treatment with Pulsed Extremely Low Frequency Electromagnetic Field (PELF-EMF) Exhibit Anti-Inflammatory and Neuroprotective Effect in Compression Spinal Cord Injury Model. Biomedicines 2022; 10:biomedicines10020325. [PMID: 35203533 PMCID: PMC8869291 DOI: 10.3390/biomedicines10020325] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 01/10/2022] [Accepted: 01/26/2022] [Indexed: 02/01/2023] Open
Abstract
Background: Spinal cord injury (SCI) pathology includes both primary and secondary events. The primary injury includes the original traumatic event, and the secondary injury, beginning immediately after the initial injury, involves progressive neuroinflammation, neuronal excitotoxicity, gliosis, and degeneration. Currently, there is no effective neuroprotective treatment for SCI. However, an accumulating body of data suggests that PELF-EMF has beneficial therapeutic effects on neurotrauma. The purpose of this study was to test the efficacy of the PELF-EMF SEQEX device using a compression SCI mouse model. Methods: C57BL/6 mice were exposed to PELF-EMF for 4 h on a daily basis for two months, beginning 2 h after a mild-moderate compression SCI. Results: The PELF-EMF treatment significantly diminished inflammatory cell infiltration and astrocyte activation by reducing Iba1, F4/80, CD68+ cells, and GAFP at the lesion borders, and increased pro-survival signaling, such as BDNF, on the neuronal cells. Moreover, the treatment exhibited a neuroprotective effect by reducing the demyelination of the axons of the white matter at the lesion’s center. Conclusions: Treatment with SEQEX demonstrated significant anti-inflammatory and neuroprotective effects. Considering our results, this safe and effective rehabilitative device, already available on the market, may provide a major therapeutic asset in the treatment of SCI.
Collapse
|
15
|
Park HJ, Choi JH, Nam MH, Seo YK. Induced Neurodifferentiation of hBM-MSCs through Activation of the ERK/CREB Pathway via Pulsed Electromagnetic Fields and Physical Stimulation Promotes Neurogenesis in Cerebral Ischemic Models. Int J Mol Sci 2022; 23:ijms23031177. [PMID: 35163096 PMCID: PMC8835447 DOI: 10.3390/ijms23031177] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 01/14/2022] [Accepted: 01/19/2022] [Indexed: 12/22/2022] Open
Abstract
Stroke is among the leading causes of death worldwide, and stroke patients are more likely to live with permanent disabilities even after treatment. Several treatments are being developed to improve the quality of life of patients; however, these treatments still have important limitations. Our study thus sought to evaluate the neural differentiation of human bone marrow mesenchymal stem cells (hBM-MSCs) at various pulsed electromagnetic field (PEMF) frequencies. Furthermore, the effects of selected frequencies in vivo were also evaluated using a mouse ischemia stroke model. Cell proliferation decreased by 20% in the PEMF group, as demonstrated by the 3-[4,5-dimethylthiazol-2-yl]-2,5-diphenyl tetrazolium bromide (MTT) assay, and lactate dehydrogenase (LDH) secretion increased by approximately 10% in an LDH release assay. Fluorescence-activated cell sorting (FACS) analysis demonstrated that CD73 and CD105 were downregulated in the PEMF group at 60 Hz. Moreover, microtubule-associated protein 2 (MAP-2) and neurofilament light chain (NF-L) were upregulated in cell cultures at 60 and 75 Hz. To assess the effects of PEMF in vivo, cerebral ischemia mice were exposed to a PEMF at 60 Hz. Neural-related proteins were significantly upregulated in the PEMF groups compared with the control and cell group. Upon conducting rotarod tests, the cell/PEMF group exhibited significant differences in motor coordination at 13 days post-treatment when compared with the control and stem-cell-treated group. Furthermore, the cell and cell/PEMF group exhibited a significant reduction in the expression of matrix metalloproteinase-9 (MMP-9), tumor necrosis factor-alpha (TNF-α) and interferon-gamma (IFN-γ) in the induced ischemic area compared with the control. Collectively, our findings demonstrated that PEMFs at 60 and 75 Hz could stimulate hBM-MSCs neural differentiation in vitro, in addition to promoting neurogenesis to enhance the functional recovery process by reducing the post-stroke inflammatory reaction.
Collapse
Affiliation(s)
| | | | | | - Young-Kwon Seo
- Correspondence: ; Tel.: +82-(31)-9615154; Fax: +82-(31)-9615521
| |
Collapse
|
16
|
Moya Gómez A, Font LP, Brône B, Bronckaers A. Electromagnetic Field as a Treatment for Cerebral Ischemic Stroke. Front Mol Biosci 2021; 8:742596. [PMID: 34557522 PMCID: PMC8453690 DOI: 10.3389/fmolb.2021.742596] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Accepted: 08/04/2021] [Indexed: 11/24/2022] Open
Abstract
Cerebral stroke is a leading cause of death and adult-acquired disability worldwide. To this date, treatment options are limited; hence, the search for new therapeutic approaches continues. Electromagnetic fields (EMFs) affect a wide variety of biological processes and accumulating evidence shows their potential as a treatment for ischemic stroke. Based on their characteristics, they can be divided into stationary, pulsed, and sinusoidal EMF. The aim of this review is to provide an extensive literature overview ranging from in vitro to even clinical studies within the field of ischemic stroke of all EMF types. A thorough comparison between EMF types and their effects is provided, as well as an overview of the signal pathways activated in cell types relevant for ischemic stroke such as neurons, microglia, astrocytes, and endothelial cells. We also discuss which steps have to be taken to improve their therapeutic efficacy in the frame of the clinical translation of this promising therapy.
Collapse
Affiliation(s)
- Amanda Moya Gómez
- UHasselt Hasselt University, BIOMED, Diepenbeek, Belgium.,Department of Biomedical Engineering, Faculty of Telecommunications, Informatics and Biomedical Engineering, Universidad de Oriente, Santiago de Cuba, Cuba
| | - Lena Pérez Font
- Centro Nacional de Electromagnetismo Aplicado, Universidad de Oriente, Santiago de Cuba, Cuba
| | - Bert Brône
- UHasselt Hasselt University, BIOMED, Diepenbeek, Belgium
| | | |
Collapse
|
17
|
Capone F, Salati S, Vincenzi F, Liberti M, Aicardi G, Apollonio F, Varani K, Cadossi R, Di Lazzaro V. Pulsed Electromagnetic Fields: A Novel Attractive Therapeutic Opportunity for Neuroprotection After Acute Cerebral Ischemia. Neuromodulation 2021; 25:1240-1247. [PMID: 34480781 DOI: 10.1111/ner.13489] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 05/18/2021] [Accepted: 06/07/2021] [Indexed: 11/30/2022]
Abstract
OBJECTIVES Acute cerebral ischemia is characterized by several pathological processes evolving during time, which contribute to the final tissue damage. Secondary processes, such as prolonged inflammatory response, impaired mitochondrial function and oxidative stress, are responsible for the progression of brain injury to the peri-infarct area, called "penumbra." Adenosine has been shown to play a crucial role in regulating the inflammatory cascade following brain ischemia. Pulsed electromagnetic fields (PEMFs) act as modulators of adenosine receptors, increasing the functionality of the endogenous adenosine. In particular, PEMF exposure induces a significant upregulation of A2A and A3 adenosine receptors in different neuronal cell types. Several lines of evidence suggest that PEMF exposure might play a neuroprotective role after ischemic damage. MATERIALS AND METHODS This review summarizes the current knowledge on the mechanism of action of PEMFs and their biological effects on neuronal damage both in preclinical and clinical studies. RESULTS PEMFs counteract hypoxia-induced apoptosis and ROS production in neuronal-like cells and exert a strong anti-inflammatory effect on microglial cells. Data from stroke animal models showed that PEMFs exposure is able to reduce the size of the infarct area and decrease the levels of pro-inflammatory mediators. In clinical studies, PEMFs stimulation proved to be safe and well tolerated. Preliminary results on acute ischemic stroke patients showed a dose-dependent reduction in the lesion size. CONCLUSIONS Altogether, these data demonstrate the efficacy of PEMFs against several mechanisms underlying ischemic damage and suggest that PEMFs might represent a novel noninvasive adjunctive treatment for acute ischemic stroke, providing neuroprotection and reducing functional deficits following ischemia.
Collapse
Affiliation(s)
- Fioravante Capone
- Unit of Neurology, Neurophysiology, and Neurobiology, Department of Medicine, Università Campus Bio-Medico, Roma, Italy
| | | | | | - Micaela Liberti
- Department of Information Engineering, Electronics and Telecommunications (DIET), University of Rome "La Sapienza", Roma, Italy
| | - Giorgio Aicardi
- Neurophysiology Research Unit, Department for Life Quality Studies, University of Bologna, Bologna, Italy
| | - Francesca Apollonio
- Department of Information Engineering, Electronics and Telecommunications (DIET), University of Rome "La Sapienza", Roma, Italy
| | - Katia Varani
- Department of Translational Medicine, University of Ferrara, Italy
| | | | - Vincenzo Di Lazzaro
- Unit of Neurology, Neurophysiology, and Neurobiology, Department of Medicine, Università Campus Bio-Medico, Roma, Italy
| |
Collapse
|
18
|
Martiñón-Gutiérrez G, Luna-Castro M, Hernández-Muñoz R. Role of insulin/glucagon ratio and cell redox state in the hyperglycaemia induced by exposure to a 60-Hz magnetic field in rats. Sci Rep 2021; 11:11666. [PMID: 34083675 PMCID: PMC8175349 DOI: 10.1038/s41598-021-91228-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2021] [Accepted: 04/29/2021] [Indexed: 11/12/2022] Open
Abstract
The exposure to extremely low-frequency electromagnetic fields (EMFs) could adversely affect the endocrine system and cellular proliferative response. Nonetheless, the use of 60-Hz EMFs in the form of magneto-therapy exerts beneficial actions on human health but can also induce hyperglycaemia. Therefore, the present study was aimed to search for metabolic responses of fed or fasted male rats to a single EMF exposure. We performed a 15 min-single exposure to 60-Hz (3.8 mT, intensity) EMF, and determined serum levels of glucose, lipids, and indicators of cellular redox state and energy parameters. A single exposure to a 60-Hz EMF induced hyperglycaemia in both animal groups, and an attenuated second serum insulin peak. The 60-Hz EMF also decreased free fatty acids and lactate serum levels, oppositely increasing pyruvate and acetoacetate levels. Significant increases in blood glucose level and rat’s glucose metabolism were related to a more oxidized cellular redox state and variations in insulin and glucagon secretion. The 60-Hz EMF’s effects were not modified in animals previously subjected to chronic EMFs exposure (14 days). In conclusion, increased serum glucose levels and glucose metabolism induced by a single 60-Hz EMF exposure were closely related to the cellular redox state and the insulin/glucagon ratio.
Collapse
Affiliation(s)
- Gabriel Martiñón-Gutiérrez
- Department of Cell Biology and Development, Institute of Cellular Physiology, Universidad Nacional Autónoma de México (UNAM), Ave. Universidad # 3000, Apdo. Postal 70-243, Coyoacán, 04510, Mexico City, Mexico
| | - María Luna-Castro
- Department of Cell Biology and Development, Institute of Cellular Physiology, Universidad Nacional Autónoma de México (UNAM), Ave. Universidad # 3000, Apdo. Postal 70-243, Coyoacán, 04510, Mexico City, Mexico
| | - Rolando Hernández-Muñoz
- Department of Cell Biology and Development, Institute of Cellular Physiology, Universidad Nacional Autónoma de México (UNAM), Ave. Universidad # 3000, Apdo. Postal 70-243, Coyoacán, 04510, Mexico City, Mexico.
| |
Collapse
|
19
|
Klimek A, Rogalska J. Extremely Low-Frequency Magnetic Field as a Stress Factor-Really Detrimental?-Insight into Literature from the Last Decade. Brain Sci 2021; 11:174. [PMID: 33572550 PMCID: PMC7912337 DOI: 10.3390/brainsci11020174] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 01/23/2021] [Accepted: 01/27/2021] [Indexed: 12/13/2022] Open
Abstract
Biological effects of extremely low-frequency magnetic field (ELF-MF) and its consequences on human health have become the subject of important and recurrent public debate. ELF-MF evokes cell/organism responses that are characteristic to a general stress reaction, thus it can be regarded as a stress factor. Exposure to ELF-MF "turns on" different intracellular mechanisms into both directions: compensatory or deleterious ones. ELF-MF can provoke morphological and physiological changes in stress-related systems, mainly nervous, hormonal, and immunological ones. This review summarizes the ELF-MF-mediated changes at various levels of the organism organization. Special attention is placed on the review of literature from the last decade. Most studies on ELF-MF effects concentrate on its negative influence, e.g., impairment of behavior towards depressive and anxiety disorders; however, in the last decade there was an increase in the number of research studies showing stimulating impact of ELF-MF on neuroplasticity and neurorehabilitation. In the face of numerous studies on the ELF-MF action, it is necessary to systematize the knowledge for a better understanding of the phenomenon, in order to reduce the risk associated with the exposure to this factor and to recognize the possibility of using it as a therapeutic agent.
Collapse
Affiliation(s)
| | - Justyna Rogalska
- Department of Animal Physiology and Neurobiology, Faculty of Biological and Veterinary Sciences, Nicolaus Copernicus University, 87-100 Torun, Poland;
| |
Collapse
|
20
|
Biermann N, Sommerauer L, Diesch S, Koch C, Jung F, Kehrer A, Prantl L, Taeger CD. The influence of pulsed electromagnetic field therapy (PEMFT) on cutaneous blood flow in healthy volunteers1. Clin Hemorheol Microcirc 2020; 76:495-501. [PMID: 33216020 DOI: 10.3233/ch-209224] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND The biophysical interaction induced by low energy pulsed electromagnetic field therapy (PEMFT) on the capillary microcirculation is not well understood. Several studies indicate a significant effect of PEMFT in patients with chronic medical conditions. OBJECTIVE The aim of this study was to evaluate the influence of PEMFT on skin microcirculation in healthy volunteers. METHODS 15 healthy participants were included. Nine PEMF treatments were applied over three weeks in an 48-hour interval. The PEMFT system (BEMER) was placed beneath one of the participants' leg with the contralateral side serving as a control. A forty-minute application period was preceded by a 10-minute resting phase. Measuring was done using two Laser Doppler probes (LEA) placed on each anterior lateral thigh. RESULTS All outcome parameters including flow, mixed venous oxygen saturation and relative venous hemoglobin showed a significant increase during the experiment when compared to the baseline values of the resting phase in both groups (p < 0.01). Comparing both groups, the measurement values during the experiment did not differ (p > 0.05) except for higher flow values in the control group (P = 0.03). Over time, baseline values of both groups showed no significant difference (p > 0.05). CONCLUSION We found a significant increase of all measurement parameters during the study compared to the baseline values with no difference between the PEMF and control group.
Collapse
Affiliation(s)
- Niklas Biermann
- Department of Plastic, Hand- and Reconstructive Surgery, University Hospital Regensburg, Regensburg, Germany
| | - Laura Sommerauer
- Department of Plastic, Hand- and Reconstructive Surgery, University Hospital Regensburg, Regensburg, Germany
| | - Sophia Diesch
- Department of Plastic, Hand- and Reconstructive Surgery, University Hospital Regensburg, Regensburg, Germany
| | - Christoph Koch
- Department of Plastic, Hand- and Reconstructive Surgery, University Hospital Regensburg, Regensburg, Germany
| | - Friedrich Jung
- Institute of Biotechnology, Molecular Cell Biology, Brandenburg University of Technology, Senftenberg, Germany
| | - Andreas Kehrer
- Department of Plastic, Hand- and Reconstructive Surgery, University Hospital Regensburg, Regensburg, Germany
| | - Lukas Prantl
- Department of Plastic, Hand- and Reconstructive Surgery, University Hospital Regensburg, Regensburg, Germany
| | - Christian D Taeger
- Department of Plastic, Hand- and Reconstructive Surgery, University Hospital Regensburg, Regensburg, Germany
| |
Collapse
|
21
|
Vincenzi F, Pasquini S, Setti S, Salati S, Cadossi R, Borea PA, Varani K. Pulsed Electromagnetic Fields Stimulate HIF-1α-Independent VEGF Release in 1321N1 Human Astrocytes Protecting Neuron-Like SH-SY5Y Cells from Oxygen-Glucose Deprivation. Int J Mol Sci 2020; 21:ijms21218053. [PMID: 33126773 PMCID: PMC7663527 DOI: 10.3390/ijms21218053] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 10/26/2020] [Accepted: 10/28/2020] [Indexed: 01/23/2023] Open
Abstract
Pulsed electromagnetic fields (PEMFs) are emerging as an innovative, non-invasive therapeutic option in different pathological conditions of the central nervous system, including cerebral ischemia. This study aimed to investigate the mechanism of action of PEMFs in an in vitro model of human astrocytes, which play a key role in the events that occur following ischemia. 1321N1 cells were exposed to PEMFs or hypoxic conditions and the release of relevant neurotrophic and angiogenic factors, such as VEGF, EPO, and TGF-β1, was evaluated by means of ELISA or AlphaLISA assays. The involvement of the transcription factor HIF-1α was studied by using the specific inhibitor chetomin and its expression was measured by flow cytometry. PEMF exposure induced a time-dependent, HIF-1α-independent release of VEGF from 1321N1 cells. Astrocyte conditioned medium derived from PEMF-exposed astrocytes significantly reduced the oxygen-glucose deprivation-induced cell proliferation and viability decrease in the neuron-like cells SH-SY5Y. These findings contribute to our understanding of PEMFs action in neuropathological conditions and further corroborate their therapeutic potential in cerebral ischemia.
Collapse
Affiliation(s)
- Fabrizio Vincenzi
- Department of Morphology, Surgery and Experimental Medicine, Section of Pharmacology, University of Ferrara, 44121 Ferrara, Italy; (S.P.); (K.V.)
- Correspondence: ; Tel.: +39-0532-455214
| | - Silvia Pasquini
- Department of Morphology, Surgery and Experimental Medicine, Section of Pharmacology, University of Ferrara, 44121 Ferrara, Italy; (S.P.); (K.V.)
| | - Stefania Setti
- Igea Biophysics Laboratory, 41012 Carpi, Italy; (S.S.); (S.S.); (R.C.)
| | - Simona Salati
- Igea Biophysics Laboratory, 41012 Carpi, Italy; (S.S.); (S.S.); (R.C.)
| | - Ruggero Cadossi
- Igea Biophysics Laboratory, 41012 Carpi, Italy; (S.S.); (S.S.); (R.C.)
| | | | - Katia Varani
- Department of Morphology, Surgery and Experimental Medicine, Section of Pharmacology, University of Ferrara, 44121 Ferrara, Italy; (S.P.); (K.V.)
| |
Collapse
|
22
|
Neuroprotective Effects of Anti-proBDNF in a Rat Photothrombotic Ischemic Model. Neuroscience 2020; 446:261-270. [PMID: 32798590 DOI: 10.1016/j.neuroscience.2020.08.008] [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: 05/10/2020] [Revised: 08/03/2020] [Accepted: 08/05/2020] [Indexed: 11/23/2022]
Abstract
Up-regulation of proBDNF in ischemic brain and the detrimental role of proBDNF on cellular survival has already been established. We propose that the up-regulated proBDNF may trigger the harmful events and evoke a secondary ischemic damage after ischemia. This study aimed to establish the neuroprotective effects of anti-proBDNF antibody in a rat photothrombotic ischemic model. Photothrombotic ischemic model was performed on Sprague Dawley rats and anti-proBDNF antibodies were administered intraperitoneally to the ischemic rats at a dose of 5 mg/kg after 6 hours (6 h) and on 3 days (3d) after ischemia. Behavioural tests were performed for sensorimotor functional analyses. Animals were euthanized at 7d for histochemical and biochemical studies. We observed higher proBDNF expression around the ischemic infarct. Higher level of apoptosis and inflammation was evident at 7d after ischemia on brain sections. Interestingly, the anti-proBDNF treatment instigated significant reduction of the infarction size as detected by Haematoxylin and Eosin (H&E) staining. Similar reduction of apoptotic signaling proteins in western blot and immunostaining after anti-proBDNF treatment was found. Up-regulation of synaptic protein expression was also observed after this treatment. Significant sensorimotor functional improvements were also noticed at 7d after anti-proBDNF treatment. We conclude that anti-proBDNF treatment is anti-apoptotic and anti-inflammatory, and plays advantageous role in promoting cellular growth and improving sensorimotor function after ischemic insult. Taken together, our study suggests that this anti-proBDNF treatment can be considered as a therapeutic approach for ischemic recovery.
Collapse
|
23
|
Zhai B, Fu J, Xiang S, Shang Y, Yan Y, Yin T, Zhang T. Repetitive transcranial magnetic stimulation ameliorates recognition memory impairment induced by hindlimb unloading in mice associated with BDNF/TrkB signaling. Neurosci Res 2020; 153:40-47. [DOI: 10.1016/j.neures.2019.04.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Revised: 02/18/2019] [Accepted: 04/08/2019] [Indexed: 02/07/2023]
|
24
|
Colella M, Camera F, Capone F, Setti S, Cadossi R, Di Lazzaro V, Apollonio F, Liberti M. Patient Semi-specific Computational Modeling of Electromagnetic Stimulation Applied to Neuroprotective Treatments in Acute Ischemic Stroke. Sci Rep 2020; 10:2945. [PMID: 32075993 PMCID: PMC7031527 DOI: 10.1038/s41598-020-59471-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Accepted: 01/13/2020] [Indexed: 11/21/2022] Open
Abstract
Neuroprotective effects of pulsed electromagnetic fields (PEMFs) have been demonstrated both in vivo and in vitro. Moreover, preliminary clinical studies have been conducted and suggested PEMFs as a possible alternative therapy to treat acute ischemic stroke. In this work, we show that it's possible to build-up a patient semi-specific head model, where the 3D reconstruction of the ischemic lesion of the patient under treatment is inserted in the head of the human body model "Duke" (v.1.0, Zurich MedTech AG). The semi-specific model will be used in the randomized, placebo-controlled, double-blind study currently ongoing. Three patients were modelled and simulated, and results showed that each ischemic lesion experiences a magnetic flux density field comparable to the one for which biological effects have been attested. Such a kind of dosimetric analysis reveals a reliable tool to assess the correlation between levels of exposure and the beneficial effect. Thus, once the on-going double blind study is complete it will prove if PEMFs treatment triggers a clinical effect, and we will then be able to characterize a dose-response curve with the methodology arranged in this study.
Collapse
Affiliation(s)
- Micol Colella
- Department of Information Engineering, Electronics and Telecommunications (DIET), University of Rome "La Sapienza", Rome, Italy
| | - Francesca Camera
- Department of Information Engineering, Electronics and Telecommunications (DIET), University of Rome "La Sapienza", Rome, Italy
| | - Fioravante Capone
- Unit of Neurology, Neurophysiology, Neurobiology, Department of Medicine, Università Campus Bio-Medico di Roma, Rome, Italy
| | | | | | - Vincenzo Di Lazzaro
- Unit of Neurology, Neurophysiology, Neurobiology, Department of Medicine, Università Campus Bio-Medico di Roma, Rome, Italy
| | - Francesca Apollonio
- Department of Information Engineering, Electronics and Telecommunications (DIET), University of Rome "La Sapienza", Rome, Italy
- Pervasive Electromagnetics Lab, University of Rome Tor Vergata, Rome, Italy
| | - Micaela Liberti
- Department of Information Engineering, Electronics and Telecommunications (DIET), University of Rome "La Sapienza", Rome, Italy.
| |
Collapse
|
25
|
Amini-Khoei H, Saghaei E, Mobini GR, Sabzevary-Ghahfarokhi M, Ahmadi R, Bagheri N, Mokhtari T. Possible involvement of PI3K/AKT/mTOR signaling pathway in the protective effect of selegiline (deprenyl) against memory impairment following ischemia reperfusion in rat. Neuropeptides 2019; 77:101942. [PMID: 31272684 DOI: 10.1016/j.npep.2019.101942] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Revised: 06/17/2019] [Accepted: 06/26/2019] [Indexed: 02/08/2023]
Abstract
Short-term cerebral ischemia led to memory dysfunction. There is a pressing need to introduce effective agents to reduce complications of the ischemia. Involvement of PI3K/AKT/mTOR signaling pathway has been determined in the neuroprotective effect of various agents. Selegiline (deprenyl) possessed neuroprotective properties. In this study global ischemia/reperfusion was established in rats. Selegiline (5 mg/kg for 7 consecutive days) administrated via intraperitoneal route. Possible involvement of PI3K/AKT/mTOR signaling pathway was evaluated using qRT-PCR, immunohistochemistry and histophatologic evaluations in the hippocampus. Spatial memory was evaluated by morris water maze (MWM). Results showed that ischemia impaired the memory and ischemic rats spent more time to find hidden platform in the MWM. Ischemia significantly decreased levels of PI3K, AKT and mTOR in the hippocampus. Histopathologic assessment revealed that the percent of dark neurons significantly increased in the CA1 area of the hippocampus of ischemic rats. Selegiline improved the memory as ischemic rats spent fewer time to find hidden platform in the MWM. Findings showed that selegiline increased the level and expression of PI3K, AKT and mTOR as well as decreased the proportion of dark neurons in the CA1 area of the pyramidal layer of the hippocampus. We concluded that selegiline, partially at least, through increases the expression of PI3K, AKT and mTOR as well as decreases the percent of dark neurons in the hippocampus could improve the memory impairment following the ischemia in rats.
Collapse
Affiliation(s)
- Hossein Amini-Khoei
- Medical Plants Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran.
| | - Elham Saghaei
- Medical Plants Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Gholam-Reza Mobini
- Medical Plants Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Milad Sabzevary-Ghahfarokhi
- Medical Plants Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Reza Ahmadi
- Medical Plants Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Nader Bagheri
- Medical Plants Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Tahmineh Mokhtari
- Nervous System Stem Cells Research Center, Semnan University of Medical Sciences, Semnan, Iran; Department of Anatomy, School of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| |
Collapse
|
26
|
Gessi S, Merighi S, Bencivenni S, Battistello E, Vincenzi F, Setti S, Cadossi M, Borea PA, Cadossi R, Varani K. Pulsed electromagnetic field and relief of hypoxia-induced neuronal cell death: The signaling pathway. J Cell Physiol 2019; 234:15089-15097. [PMID: 30656694 DOI: 10.1002/jcp.28149] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Accepted: 01/02/2019] [Indexed: 01/24/2023]
Abstract
Low-energy low-frequency pulsed electromagnetic fields (PEMFs) exert several protective effects, such as the regulation of kinases, transcription factors as well as cell viability in both central and peripheral biological systems. However, it is not clear on which bases they affect neuroprotection and the mechanism responsible is yet unknown. In this study, we have characterized in nerve growth factor-differentiated pheochromocytoma PC12 cells injured with hypoxia: (i) the effects of PEMF exposure on cell vitality; (ii) the protective pathways activated by PEMFs to relief neuronal cell death, including adenylyl cyclase, phospholipase C, protein kinase C epsilon and delta, p38, ERK1/2, JNK1/2 mitogen-activated protein kinases, Akt and caspase-3; (iii) the regulation by PEMFs of prosurvival heat-shock proteins of 70 (HSP70), cAMP response element-binding protein (CREB), brain-derived neurotrophic factor (BDNF), and Bcl-2 family proteins. The results obtained in this study show a protective effect of PEMFs that are able to reduce neuronal cell death induced by hypoxia by modulating p38, HSP70, CREB, BDNF, and Bcl-2 family proteins. Specifically, we found a rapid activation (30 min) of p38 kinase cascade, which in turns enrolles HSP70 survival chaperone molecule, resulting in a significant CREB phosphorylation increase (24 hr). In this cascade, later (48 hr), BDNF and the antiapoptotic pathway regulated by the Bcl-2 family of proteins are recruited by PEMFs to enhance neuronal survival. This study paves the way to elucidate the mechanisms triggered by PEMFs to act as a new neuroprotective approach to treat cerebral ischemia by reducing neuronal cell death.
Collapse
Affiliation(s)
- Stefania Gessi
- Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| | - Stefania Merighi
- Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| | - Serena Bencivenni
- Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| | | | - Fabrizio Vincenzi
- Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| | | | | | - Pier Andrea Borea
- Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| | | | - Katia Varani
- Department of Medical Sciences, University of Ferrara, Ferrara, Italy.,University Center for Studies on Gender Medicine, University of Ferrara, Ferrara, Italy
| |
Collapse
|
27
|
Signaling pathways involved in anti-inflammatory effects of Pulsed Electromagnetic Field in microglial cells. Cytokine 2019; 125:154777. [PMID: 31400640 DOI: 10.1016/j.cyto.2019.154777] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Revised: 06/14/2019] [Accepted: 07/09/2019] [Indexed: 11/27/2022]
Abstract
Literature studies suggest important protective effects of low-frequency, low-energy pulsed electromagnetic fields (PEMFs) on inflammatory pathways affecting joint and cerebral diseases. However, it is not clear on which bases they affect neuroprotection and the mechanism responsible is yet unknown. Therefore the aim of this study was to identify the molecular targets of PEMFs anti-neuroinflammatory action. The effects of PEMF exposure in cytokine production by lipopolysaccharide (LPS)-activated N9 microglial cells as well as the pathways involved, including adenylyl cyclase (AC), phospholipase C (PLC), protein kinase C epsilon (PKC-ε) and delta (PKC-δ), p38, ERK1/2, JNK1/2 mitogen activated protein kinases (MAPK), Akt and caspase 1, were investigated. In addition, the ability of PEMFs to modulate ROS generation, cell invasion and phagocytosis, was addressed. PEMFs reduced the LPS-increased production of TNF-α and IL-1β in N9 cells, through a pathway involving JNK1/2. Furthermore, they decreased the LPS-induced release of IL-6, by a mechanism not dependent on AC, PLC, PKC-ε, PKC-δ, p38, ERK1/2, JNK1/2, Akt and caspase 1. Importantly, a significant effect of PEMFs in the reduction of crucial cell functions specific of microglia like ROS generation, cell invasion and phagocytosis was found. PEMFs inhibit neuroinflammation in N9 cells through a mechanism involving, at least in part, the activation of JNK MAPK signalling pathway and may be relevant to treat a variety of diseases characterized by neuroinflammation.
Collapse
|
28
|
Consales C, Panatta M, Butera A, Filomeni G, Merla C, Carrì MT, Marino C, Benassi B. 50-Hz magnetic field impairs the expression of iron-related genes in the in vitro SOD1 G93A model of amyotrophic lateral sclerosis. Int J Radiat Biol 2019; 95:368-377. [PMID: 30513241 DOI: 10.1080/09553002.2019.1552378] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
PURPOSE We characterized the response to the extremely low frequency magnetic field (ELF-MF) in an in vitro model of familial Amyotrophic Lateral Sclerosis (fALS), carrying two mutant variants of the superoxide dismutase 1 (SOD1) gene. MATERIALS AND METHODS SH-SY5Y human neuroblastoma cells, stably over-expressing the wild type, the G93A or the H46R mutant SOD1 cDNA, were exposed to either the ELF-MF (50 Hz, 1 mT) or the sham control field, up to 72 h. Analysis of (i) viability, proliferation and apoptosis, (ii) reactive oxygen species generation, and (iii) assessment of the iron metabolism, were carried out in all clones in response to the MF exposure. RESULTS We report that 50-Hz MF exposure induces: (i) no change in proliferation and viability; (ii) no modulation of the intracellular superoxide and H2O2 levels; (iii) a significant deregulation in the expression of iron-related genes IRP1, MFRN1 and TfR1, this evidence being exclusive for the SOD1G93A clone and associated with a slight (p = .0512) difference in the total iron content. CONCLUSIONS 50-Hz MF affects iron homeostasis in the in vitro SOD1G93A ALS model.
Collapse
Affiliation(s)
- Claudia Consales
- a Department of Energy and Sustainable Economic Development , Division of Health Protection Technologies, ENEA-Italian National Agency for New Technologies , Rome , Italy
| | - Martina Panatta
- a Department of Energy and Sustainable Economic Development , Division of Health Protection Technologies, ENEA-Italian National Agency for New Technologies , Rome , Italy.,b Department of Chemistry and Biochemistry , University of Bern , Bern , Switzerland
| | - Alessio Butera
- a Department of Energy and Sustainable Economic Development , Division of Health Protection Technologies, ENEA-Italian National Agency for New Technologies , Rome , Italy
| | - Giuseppe Filomeni
- c Department of Biology , University of Rome Tor Vergata , Rome , Italy.,d Cell Stress and Survival Unit, Center for Autophagy, Recycling and Disease (CARD) , Danish Cancer Society Research Center , Copenhagen , Denmark
| | - Caterina Merla
- a Department of Energy and Sustainable Economic Development , Division of Health Protection Technologies, ENEA-Italian National Agency for New Technologies , Rome , Italy
| | | | - Carmela Marino
- a Department of Energy and Sustainable Economic Development , Division of Health Protection Technologies, ENEA-Italian National Agency for New Technologies , Rome , Italy
| | - Barbara Benassi
- a Department of Energy and Sustainable Economic Development , Division of Health Protection Technologies, ENEA-Italian National Agency for New Technologies , Rome , Italy
| |
Collapse
|
29
|
Wang C, Liu Y, Wang Y, Wei Z, Suo D, Ning G, Wu Q, Feng S, Wan C. Low‑frequency pulsed electromagnetic field promotes functional recovery, reduces inflammation and oxidative stress, and enhances HSP70 expression following spinal cord injury. Mol Med Rep 2019; 19:1687-1693. [PMID: 30628673 PMCID: PMC6390012 DOI: 10.3892/mmr.2019.9820] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Accepted: 11/23/2018] [Indexed: 12/18/2022] Open
Abstract
Low-frequency pulsed electromagnetic fields (LPEMFs) have been reported to be protective for multiple diseases. However, whether the administration of LPEMFs inhibits inflammation and oxidative stress following spinal cord injury requires further investigation. In the current study, a contusion spinal cord injury model was used and LPEMFs administration was applied to investigate the molecular changes, including inflammation, oxidative stress and heat shock protein 70 (HSP70) levels. The results revealed that LPEMFs significantly promoted functional recovery following spinal cord injury, as demonstrated by an increased Basso, Beattie and Bresnahan score. The results demonstrated that LPEMFs decreased the expression of inflammatory factors, including tumor necrosis factor-α, interleukin-1β and nuclear factor-κB. Additionally, LPEMFs exposure reduced the levels of inducible nitric oxide synthase and reactive oxygen species, and upregulated the expression of catalase and superoxide dismutase. Furthermore, treatment with LPEMFs significantly enhanced the expression of HSP70 in spinal cord-injured rats. Overall, the present study revealed that LPEMFs promote functional recovery following spinal cord injury, potentially by modulating inflammation, oxidative stress and HSP70.
Collapse
Affiliation(s)
- Chunyan Wang
- Department of Rehabilitation Medicine, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| | - Yang Liu
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| | - Yao Wang
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| | - Zhijian Wei
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| | - Dongmei Suo
- Department of Rehabilitation Medicine, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| | - Guangzhi Ning
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| | - Qiuli Wu
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| | - Shiqing Feng
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| | - Chunxiao Wan
- Department of Rehabilitation Medicine, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| |
Collapse
|
30
|
Cichoń N, Bijak M, Czarny P, Miller E, Synowiec E, Sliwinski T, Saluk-Bijak J. Increase in Blood Levels of Growth Factors Involved in the Neuroplasticity Process by Using an Extremely Low Frequency Electromagnetic Field in Post-stroke Patients. Front Aging Neurosci 2018; 10:294. [PMID: 30319398 PMCID: PMC6168626 DOI: 10.3389/fnagi.2018.00294] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Accepted: 09/06/2018] [Indexed: 12/18/2022] Open
Abstract
Background: Neuroplasticity ensures the improvement of functional status in patients after stroke. The aim of this study was to evaluate the effect of extremely low-frequency electromagnetic field therapy (ELF-EMF) on brain plasticity in the rehabilitation of patients after stroke. Methods: Forty-eight patients were divided into two groups underwent the same rehabilitation program, but in the study group, the patients additionally were exposed to a standard series of 10 ELF-EMF treatments. To determine the level of neuroplasticity, we measured the plasma level of the brain-derived neurotrophic factor (BDNF), the vascular-endothelial growth factor, as well as BDNF mRNA expression. Additionally, we determined the molecule levels for hepatocyte growth factor, stem cell factor, stromal cell-derived factor 1α, nerve growth factor β, and leukemia inhibitory factor, using 5plex cytokine panel in plasma. After 4 weeks, during which patients had undergone neurorehabilitation and neurological examinations, we assessed functional recovery using the Barthel Index, Mini-Mental State Examination (MMSE), Geriatric Depression Scale, National Institutes of Health Stroke Scale (NIHSS), and the modified Rankin Scale (mRS). Results: We observed that ELF-EMF significantly increased growth factors and cytokine levels involved in neuroplasticity, as well as promoted an enhancement of functional recovery in post-stroke patients. Additionally, we presented evidence that these effects could be related to the increase of gene expression on the mRNA level. Moreover, a change of BDNF plasma level was positively correlated with the Barthel Index, MMSE, and negatively correlated with GDS. Conclusion: Extremely low-frequency electromagnetic field therapy improves the effectiveness of rehabilitation of post-stroke patients by improving neuroplasticity processes.
Collapse
Affiliation(s)
- Natalia Cichoń
- Department of General Biochemistry, Faculty of Biology and Environmental Protection, University of Lodz, Łódź, Poland
| | - Michał Bijak
- Department of General Biochemistry, Faculty of Biology and Environmental Protection, University of Lodz, Łódź, Poland
| | - Piotr Czarny
- Department of Medical Biochemistry, Medical University of Lodz, Łódź, Poland
| | - Elżbieta Miller
- Department of Physical Medicine, Medical University of Lodz, Łódź, Poland.,Neurorehabilitation Ward, III General Hospital in Lodz, Łódź, Poland
| | - Ewelina Synowiec
- Laboratory of Medical Genetics, Department of Molecular Genetics, Faculty of Biology and Environmental Protection, University of Lodz, Łódź, Poland
| | - Tomasz Sliwinski
- Laboratory of Medical Genetics, Department of Molecular Genetics, Faculty of Biology and Environmental Protection, University of Lodz, Łódź, Poland
| | - Joanna Saluk-Bijak
- Department of General Biochemistry, Faculty of Biology and Environmental Protection, University of Lodz, Łódź, Poland
| |
Collapse
|
31
|
Zidan N, Fenn J, Griffith E, Early PJ, Mariani CL, Muñana KR, Guevar J, Olby NJ. The Effect of Electromagnetic Fields on Post-Operative Pain and Locomotor Recovery in Dogs with Acute, Severe Thoracolumbar Intervertebral Disc Extrusion: A Randomized Placebo-Controlled, Prospective Clinical Trial. J Neurotrauma 2018; 35:1726-1736. [DOI: 10.1089/neu.2017.5485] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
Affiliation(s)
- Natalia Zidan
- Department of Clinical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina
| | - Joe Fenn
- Department of Clinical Science and Services, Royal Veterinary College, Hatfield, United Kingdom
| | - Emily Griffith
- Department of Statistics, North Carolina State University, Raleigh, North Carolina
| | - Peter J. Early
- Department of Clinical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina
| | - Chris L. Mariani
- Department of Clinical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina
- Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina
| | - Karen R. Muñana
- Department of Clinical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina
- Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina
| | - Julien Guevar
- Department of Clinical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina
| | - Natasha J. Olby
- Department of Clinical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina
- Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina
| |
Collapse
|
32
|
Premi E, Benussi A, La Gatta A, Visconti S, Costa A, Gilberti N, Cantoni V, Padovani A, Borroni B, Magoni M. Modulation of long-term potentiation-like cortical plasticity in the healthy brain with low frequency-pulsed electromagnetic fields. BMC Neurosci 2018; 19:34. [PMID: 29895259 PMCID: PMC5998451 DOI: 10.1186/s12868-018-0434-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2017] [Accepted: 05/29/2018] [Indexed: 02/04/2023] Open
Abstract
Background Non-depolarizing magnetic fields, like low frequency-pulsed electromagnetic fields (LF-PEMFs) have shown the ability to modulate living structures, principally by influencing synaptic activity and ion channels on cellular membranes. Recently, the CTU Mega 20 device was presented as a molecular accelerator, using energy up to 200 J and providing high-power (2 Tesla) pulsating fields with a water-repulsive (diamagnetic) action and tissue biostimulation. We tested the hypothesis that LF-PEMFs could modulate long-term corticospinal excitability in healthy brains by applying CTU Mega 20®. Ten healthy subjects without known neurological and/or psychiatric diseases entered the study. A randomized double-blind sham-controlled crossover design was employed, recording TMS parameters (amplitude variation of the motor evoked potential as index of cortical excitability perturbations of the motor system) before (pre) and after (post + 0, + 15, + 30 min) a single CTU Mega 20 session on the corresponding primary right-hand motor area, using a real (magnetic field = 2 Tesla; intensity = 90 J; impulse frequency = 7 Hz; duration = 15 min) or sham device. A two-way repeated measures ANOVA with TIME (pre, post + 0, + 15, + 30 min) and TREATMENT (real vs. sham stimulation) as within-subjects factor was applied. Results A significant TIME × TREATMENT interaction was found (p < 0.001). Post hoc comparisons showed a significant effect of TIME, with significant differences at + 0, + 15 and + 30 min compared to baseline after real stimulation (all p < 0.05) but not after sham stimulation (all p < 0.05) and significant effects of TREATMENT, with significant differences at + 0, + 15 and + 30 min for real stimulation compared to sham stimulation (all p < 0.005). No significant depolarizing effects were detected throughout the (real) stimulation. Conclusions Our proof-of-concept study in healthy subjects supports the idea that non-ionizing LF-PEMFs induced by the CTU Mega 20 diamagnetic acceleration system could represent a new approach for brain neuromodulation. Further studies to optimize protocol parameters for different neurological and psychiatric conditions are warranted. Trial Registration The present work has been retrospectively registered as clinical trial on ClinicalTrials.gov NCT03537469 and publicly released on May 24, 2018 Electronic supplementary material The online version of this article (10.1186/s12868-018-0434-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Enrico Premi
- Stroke Unit, Azienda Socio Sanitaria Territoriale "Spedali Civili", "Spedali Civili" Hospital, Piazza Spedali Civili 1, 25123, Brescia, Italy. .,Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy.
| | - Alberto Benussi
- Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | | | - Stefano Visconti
- Rehabilitation Unit, Casa di Cura "Villa Barbarano", Salò, Brescia, Italy
| | - Angelo Costa
- Stroke Unit, Azienda Socio Sanitaria Territoriale "Spedali Civili", "Spedali Civili" Hospital, Piazza Spedali Civili 1, 25123, Brescia, Italy
| | - Nicola Gilberti
- Stroke Unit, Azienda Socio Sanitaria Territoriale "Spedali Civili", "Spedali Civili" Hospital, Piazza Spedali Civili 1, 25123, Brescia, Italy
| | - Valentina Cantoni
- Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Alessandro Padovani
- Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Barbara Borroni
- Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Mauro Magoni
- Stroke Unit, Azienda Socio Sanitaria Territoriale "Spedali Civili", "Spedali Civili" Hospital, Piazza Spedali Civili 1, 25123, Brescia, Italy
| |
Collapse
|
33
|
Photothrombotic Stroke as a Model of Ischemic Stroke. Transl Stroke Res 2017; 9:437-451. [DOI: 10.1007/s12975-017-0593-8] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Revised: 11/14/2017] [Accepted: 11/24/2017] [Indexed: 12/20/2022]
|