1
|
Sun S, Bok J, Jang Y, Seo H. Brain Disease-Modifying Effects of Radiofrequency as a Non-Contact Neuronal Stimulation Technology. Int J Mol Sci 2025; 26:2268. [PMID: 40076887 PMCID: PMC11900402 DOI: 10.3390/ijms26052268] [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: 12/30/2024] [Revised: 02/05/2025] [Accepted: 02/06/2025] [Indexed: 03/14/2025] Open
Abstract
Non-invasive, non-contact, and painless methods of electrical stimulation to enhance neural function have been widely studied in recent years, particularly in the context of neurodegenerative diseases such as Alzheimer's disease (AD) and related dementias, which cause cognitive decline and other neurological symptoms. Radiofrequency (RF), which is a rate of oscillation in the range of 3 kHz to 300 GHz (3 THz), has been suggested as one potential non-contact neuronal stimulation (NCNS) technique for improving brain function. A new type of electrical stimulation uses a radiofrequency electromagnetic field (RF-EMF). RF exposure has been shown to modulate neural stimulation and influence various brain activities in in vitro and in vivo models. Recent studies have explored the effects of RF-EMF on human physiology, particularly in areas such as brain activity, cognition, and sleep behavior. In this review, we summarize recent findings about the effects of non-contact stimulations in in vitro studies, in vivo animal models, and human clinical cases.
Collapse
Affiliation(s)
- Shulei Sun
- Department of Molecular and Life Sciences, Institute for Precision Therapeutics, Center for Bionano Intelligence Education and Research, Hanyang University, 55 Hanyangdaehak-ro, Ansan 15588, Republic of Korea
| | - Junsoo Bok
- Department of Medical and Digital Engineering, College of Engineering, Hanyang University, Seoul 04736, Republic of Korea
| | - Yongwoo Jang
- Department of Medical and Digital Engineering, College of Engineering, Hanyang University, Seoul 04736, Republic of Korea
- Department of Pharmacology, College of Medicine, Hanyang University, Seoul 04736, Republic of Korea
| | - Hyemyung Seo
- Department of Molecular and Life Sciences, Institute for Precision Therapeutics, Center for Bionano Intelligence Education and Research, Hanyang University, 55 Hanyangdaehak-ro, Ansan 15588, Republic of Korea
| |
Collapse
|
2
|
Baranowski R, Amschler J, Wittwer D, Arendash GW. Memory enhancement by transcranial radiofrequency wave treatment occurs without appreciably increasing brain temperature. Phys Eng Sci Med 2025; 48:239-250. [PMID: 39760845 DOI: 10.1007/s13246-024-01508-8] [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: 09/19/2024] [Accepted: 12/04/2024] [Indexed: 01/07/2025]
Abstract
We have previously shown in small studies that full brain Transcranial Radiofrequency Wave Treatment (TRFT) to subjects with Alzheimer's Disease could stop and reverse their cognitive decline. An 8-emitter head device, the "MemorEM", was used in these studies to provide TRFT at 915 MHz frequency and power level of 1.6 W/kg Specific Absorption Rate (SAR) during daily 1-hour treatments. Although no deleterious side effects during up to 2.5 years of treatment were reported, it is important to rule out the possibility that brain heating will occur during TRFT in humans at a higher power level of 4.0 W/kg SAR, which is anticipated for future clinical testing in order to increase treatment intensity/efficacy to deep sub-cortical areas. To examine if brain heating occurs during a single 1-hour treatment at 4 W/kg SAR, a hollow human head phantom filled with brain-analogous gel and with an attached MemorEM head device was utilized. Brain temperatures were taken at 64 specific coordinates within the brain gel before and immediately following one-hour of TRFT. Results revealed none of the 64 sites having a temperature increase after TRFT of 1 °C or more. Indeed, 45 of the 64 sites exhibited a temperature rise of less than 0.5 °C, with just three sites exhibiting an increase between 0.75 and 0.9 °C. These results demonstrate that TRFT in a human head phantom that mimics the electromagnetic properties of the human head, does not appreciably increase brain temperature (i.e., is non-thermal) at 915 MHz frequency and 4 W/kg SAR power level. Thus, TRFT would appear to be safe at 4 W/kg for long-term daily treatments.
Collapse
Affiliation(s)
| | | | | | - Gary W Arendash
- RF Longevity, 428 E. Thunderbird Road, Phoenix, SE, AZ, 85022, USA.
- NeuroEM Therapeutics, 501 E. Kennedy Blvd, Tampa, FL, 33602, USA.
| |
Collapse
|
3
|
Perez FP, Walker B, Morisaki J, Kanakri H, Rizkalla M. Neurostimulation devices to treat Alzheimer's disease. EXPLORATION OF NEUROSCIENCE 2025; 4:100674. [PMID: 40084342 PMCID: PMC11904933 DOI: 10.37349/en.2025.100674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/01/2024] [Accepted: 02/14/2025] [Indexed: 03/16/2025]
Abstract
The use of neurostimulation devices for the treatment of Alzheimer's disease (AD) is a growing field. In this review, we examine the mechanism of action and therapeutic indications of these neurostimulation devices in the AD process. Rapid advancements in neurostimulation technologies are providing non-pharmacological relief to patients affected by AD pathology. Neurostimulation therapies include electrical stimulation that targets the circuitry-level connection in important brain areas such as the hippocampus to induce therapeutic neuromodulation of dysfunctional neural circuitry and electromagnetic field (EMF) stimulation that targets anti-amyloid molecular pathways to promote the degradation of beta-amyloid (Aβ). These devices target specific or diffuse cortical and subcortical brain areas to modulate neuronal activity at the electrophysiological or molecular pathway level, providing therapeutic effects for AD. This review attempts to determine the most effective and safe neurostimulation device for AD and provides an overview of potential and current clinical indications. Several EMF devices have shown a beneficial or harmful effect in cell cultures and animal models but not in AD human studies. These contradictory results may be related to the stimulation parameters of these devices, such as frequency, penetration depth, power deposition measured by specific absorption rate, time of exposure, type of cell, and tissue dielectric properties. Based on this, determining the optimal stimulation parameters for EMF devices in AD and understanding their mechanism of action is essential to promote their clinical application, our review suggests that repeated EMF stimulation (REMFS) is the most appropriate device for human AD treatments. Before its clinical application, it is necessary to consider the complicated and interconnected genetic and epigenetic effects of REMFS-biological system interaction. This will move forward the urgently needed therapy of EMF in human AD.
Collapse
Affiliation(s)
- Felipe P. Perez
- Department of Medicine, Division of General Internal Medicine and Geriatrics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Brett Walker
- Department of Medicine, Division of General Internal Medicine and Geriatrics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Jorge Morisaki
- Department of Bioengineering, University of Illinois at Chicago, Chicago, IL 60607, USA
| | - Haitham Kanakri
- Department of Electrical and Computer Engineering, Purdue University, Indianapolis, IN 46202, USA
| | - Maher Rizkalla
- Department of Electrical and Computer Engineering, Purdue University, Indianapolis, IN 46202, USA
| |
Collapse
|
4
|
Arendash GW. The Brain Toxin Cleansing of Sleep Achieved During Wakefulness. J Clin Med 2025; 14:926. [PMID: 39941597 PMCID: PMC11818883 DOI: 10.3390/jcm14030926] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 01/21/2025] [Accepted: 01/23/2025] [Indexed: 02/16/2025] Open
Abstract
A primary purpose of sleep for humans is to remove toxins and metabolic wastes from the brain (e.g., Aβ, tau, lactate) that would otherwise build up and compromise brain functionality. There are currently no drugs or devices that have been clinically shown in humans to enhance brain toxin removal, either during sleep or wakefulness. This perspective article focuses on a recently (re)discovered major route of toxin drainage from the human brain through meningeal lymphatic vessels (mLVs) and the primary enhancer of their flow-the cytokine Vascular Endothelial Growth Factor (VEGF). The purpose of this perspective article is to present pre-clinical and clinical evidence relevant to a new bioengineered technology (Transcranial Radiofrequency Treatment; TRFT) that appears to enhance mLV flow to increase brain toxin cleansing in humans during wakefulness. In being both safe and non-invasive, TRFT is administered in-home, presently through a device called "MemorEM". Two months of daily TRFT during wakefulness increased the typically low plasma/brain levels of VEGF in Alzheimer's Disease (AD) subjects, which was associated with increased Aβ and tau toxin removal from their brains during wakefulness-ostensibly through VEGF-increased mLV flow. Even irrespective of baseline VEGF levels, brain toxin cleansing was increased by TRFT in AD subjects, who also experienced a notable reversal of their cognitive impairment after TRFT. Additional clinical studies are nonetheless required to firmly establish TRFT's brain cleansing abilities during wakefulness. In performing a major duty of sleep, TRFT during wakefulness is proposed as a viable intervention to counter the decline in nighttime brain toxin cleansing that occurs with aging and in multiple brain diseases, most notably Alzheimer's Disease. The implications of TRFT for insomnia and for sleep deprivation are also discussed, as is the potential for TRFT to extend healthy human longevity.
Collapse
Affiliation(s)
- Gary W Arendash
- RF Longevity, 428 E. Thunderbird Rd., Suite 431, Phoenix, AZ 85022, USA
| |
Collapse
|
5
|
Teranishi M, Ito M, Huang Z, Nishiyama Y, Masuda A, Mino H, Tachibana M, Inada T, Ohno K. Extremely Low-Frequency Electromagnetic Field (ELF-EMF) Increases Mitochondrial Electron Transport Chain Activities and Ameliorates Depressive Behaviors in Mice. Int J Mol Sci 2024; 25:11315. [PMID: 39457098 PMCID: PMC11508854 DOI: 10.3390/ijms252011315] [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: 09/08/2024] [Revised: 10/17/2024] [Accepted: 10/17/2024] [Indexed: 10/28/2024] Open
Abstract
Compromised mitochondrial electron transport chain (ETC) activities are associated with depression in humans and rodents. However, the effects of the enhancement of mitochondrial ETC activities on depression remain elusive. We recently reported that an extremely low-frequency electromagnetic field (ELF-EMF) of as low as 10 μT induced hormetic activation of mitochondrial ETC complexes in human/mouse cultured cells and mouse livers. Chronic social defeat stress (CSDS) for 10 consecutive days caused behavioral defects mimicking depression in mice, and using an ELF-EMF for two to six weeks ameliorated them. CSDS variably decreased the mitochondrial ETC proteins in the prefrontal cortex (PFC) in 10 days, which were increased by an ELF-EMF in six weeks. CSDS had no effect on the mitochondrial oxygen consumption rate in the PFC in 10 days, but using an ELF-EMF for six weeks enhanced it. CSDS inactivated SOD2 by enhancing its acetylation and increased lipid peroxidation in the PFC. In contrast, the ELF-EMF activated the Sirt3-FoxO3a-SOD2 pathway and suppressed lipid peroxidation. Furthermore, CSDS increased markers for mitophagy, which was suppressed by the ELF-EMF in six weeks. The ELF-EMF exerted beneficial hormetic effects on mitochondrial energy production, mitochondrial antioxidation, and mitochondrial dynamics in a mouse model of depression. We envisage that an ELF-EMF is a promising therapeutic option for depression.
Collapse
Affiliation(s)
- Masaki Teranishi
- Division of Neurogenetics, Center for Neurological Diseases and Cancer, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan; (M.T.); (Z.H.); (Y.N.); (A.M.)
| | - Mikako Ito
- Division of Neurogenetics, Center for Neurological Diseases and Cancer, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan; (M.T.); (Z.H.); (Y.N.); (A.M.)
| | - Zhizhou Huang
- Division of Neurogenetics, Center for Neurological Diseases and Cancer, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan; (M.T.); (Z.H.); (Y.N.); (A.M.)
| | - Yuki Nishiyama
- Division of Neurogenetics, Center for Neurological Diseases and Cancer, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan; (M.T.); (Z.H.); (Y.N.); (A.M.)
| | - Akio Masuda
- Division of Neurogenetics, Center for Neurological Diseases and Cancer, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan; (M.T.); (Z.H.); (Y.N.); (A.M.)
| | - Hiroyuki Mino
- Division of Material Science (Physics), Graduate School of Science, Nagoya University, Nagoya 464-8602, Japan;
| | - Masako Tachibana
- Department of Psychiatry, Nagoya University Hospital, Nagoya 466-8560, Japan;
| | - Toshiya Inada
- Department of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan;
| | - Kinji Ohno
- Division of Neurogenetics, Center for Neurological Diseases and Cancer, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan; (M.T.); (Z.H.); (Y.N.); (A.M.)
- Graduate School of Nutritional Sciences, Nagoya University of Arts and Sciences, Nisshin 470-0196, Japan
| |
Collapse
|
6
|
Bontempi B, Lévêque P, Dubreuil D, Jay TM, Edeline JM. Effects of Head-Only Exposure to 900 MHz GSM Electromagnetic Fields in Rats: Changes in Neuronal Activity as Revealed by c-Fos Imaging without Concomitant Cognitive Impairments. Biomedicines 2024; 12:1954. [PMID: 39335468 PMCID: PMC11428239 DOI: 10.3390/biomedicines12091954] [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: 07/18/2024] [Revised: 08/07/2024] [Accepted: 08/22/2024] [Indexed: 09/30/2024] Open
Abstract
Over the last two decades, animal models have been used to evaluate the physiological and cognitive effects of mobile phone exposure. Here, we used a head-only exposure system in rats to determine whether exposure to 900 MHz GSM electromagnetic fields (EMFs) induces regional changes in neuronal activation as revealed by c-Fos imaging. In a first study, rats were exposed for 2 h to brain average specific absorption rates (BASARs) ranging from 0.5 to 6 W/kg. Changes in neuronal activation were found to be dose-dependent, with significant increases in c-Fos expression occurring at BASAR of 1 W/kg in prelimbic, infralimbic, frontal, and cingulate cortices. In a second study, rats were submitted to either a spatial working memory (WM) task in a radial maze or a spatial reference memory (RM) task in an open field arena. Exposures (45 min) were conducted before each daily training session (BASARs of 1 and 3.5 W/kg). Control groups included sham-exposed and control cage animals. In both tasks, behavioral performance evolved similarly in the four groups over testing days. However, c-Fos staining was significantly reduced in cortical areas (prelimbic, infralimbic, frontal, cingulate, and visual cortices) and in the hippocampus of animals engaged in the WM task (BASARs of 1 and 3.5 W/kg). In the RM task, EMF exposure-induced decreases were limited to temporal and visual cortices (BASAR of 1 W/kg). These results demonstrate that both acute and subchronic exposures to 900 MHz EMFs can produce region-specific changes in brain activity patterns, which are, however, insufficient to induce detectable cognitive deficits in the behavioral paradigms used here.
Collapse
Affiliation(s)
- Bruno Bontempi
- Institut de Neurosciences Cognitives et Intégratives d'Aquitaine, CNRS UMR 5287, Université de Bordeaux et Ecole Pratique des Hautes Etudes, 33000 Bordeaux, France
| | - Philippe Lévêque
- XLIM, CNRS UMR 6172, Université de Limoges, 87060 Limoges, France
| | - Diane Dubreuil
- Institut des Neurosciences Paris-Saclay (NeuroPSI), Université Paris-Saclay, CNRS, CEA Paris-Saclay, bât 151, 91400 Saclay, France
| | - Thérèse M Jay
- Institut de Psychiatrie et Neurosciences de Paris, UMR_S 1266 INSERM, Université Paris Cité, 75014 Paris, France
| | - Jean-Marc Edeline
- Institut des Neurosciences Paris-Saclay (NeuroPSI), Université Paris-Saclay, CNRS, CEA Paris-Saclay, bât 151, 91400 Saclay, France
| |
Collapse
|
7
|
Branigan KS, Dotta BT. Cognitive Decline: Current Intervention Strategies and Integrative Therapeutic Approaches for Alzheimer's Disease. Brain Sci 2024; 14:298. [PMID: 38671950 PMCID: PMC11048559 DOI: 10.3390/brainsci14040298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2024] [Revised: 03/14/2024] [Accepted: 03/18/2024] [Indexed: 04/28/2024] Open
Abstract
Alzheimer's disease (AD) represents a pressing global health challenge, with an anticipated surge in diagnoses over the next two decades. This progressive neurodegenerative disorder unfolds gradually, with observable symptoms emerging after two decades of imperceptible brain changes. While traditional therapeutic approaches, such as medication and cognitive therapy, remain standard in AD management, their limitations prompt exploration into novel integrative therapeutic approaches. Recent advancements in AD research focus on entraining gamma waves through innovative methods, such as light flickering and electromagnetic fields (EMF) stimulation. Flickering light stimulation (FLS) at 40 Hz has demonstrated significant reductions in AD pathologies in both mice and humans, providing improved cognitive functioning. Additionally, recent experiments have demonstrated that APOE mutations in mouse models substantially reduce tau pathologies, with microglial modulation playing a crucial role. EMFs have also been demonstrated to modulate microglia. The exploration of EMFs as a therapeutic approach is gaining significance, as many recent studies have showcased their potential to influence microglial responses. Th article concludes by speculating on the future directions of AD research, emphasizing the importance of ongoing efforts in understanding the complexities of AD pathogenesis through a holistic approach and developing interventions that hold promise for improved patient outcomes.
Collapse
Affiliation(s)
| | - Blake T. Dotta
- Behavioural Neuroscience & Biology Programs, School of Natural Science, Laurentian University, Sudbury, ON P3E2C6, Canada
| |
Collapse
|
8
|
Sleem T, Decourt B, Sabbagh MN. Nonmedication Devices in Development for the Treatment of Alzheimer's Disease. J Alzheimers Dis Rep 2024; 8:241-255. [PMID: 38405349 PMCID: PMC10894612 DOI: 10.3233/adr-230115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Accepted: 01/13/2024] [Indexed: 02/27/2024] Open
Abstract
Huge investments continue to be made in treatment for Alzheimer's disease (AD), with more than one hundred drugs currently in development. Pharmacological approaches and drug development, particularly those targeting amyloid-β, have dominated the therapeutic landscape. At the same time, there is also a growing interest in devices for treating AD. This review aimed to identify and describe devices under development for AD treatment. In this review, we queried the devices that are in development for the treatment of AD. PubMed was searched through the end of 2021 using the terms "device," "therapeutics," and "Alzheimer's" for articles that report on devices to treat AD. Ten devices with 31 references were identified as actively being developed for the treatment of AD. Many of these devices are far along in development. Device-based therapies are often overlooked when evaluating treatment approaches to AD. However, many devices for treating AD are in development and some show promising results.
Collapse
Affiliation(s)
- Tamara Sleem
- Department of Neurology, Barrow Neurological Institute, St. Joseph’s Hospital and Medical Center, Phoenix, AZ, USA
| | - Boris Decourt
- Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, School of Medicine, Lubbock, TX, USA
| | - Marwan N. Sabbagh
- Department of Neurology, Barrow Neurological Institute, St. Joseph’s Hospital and Medical Center, Phoenix, AZ, USA
| |
Collapse
|
9
|
Nystuen KL, McNamee SM, Akula M, Holton KM, DeAngelis MM, Haider NB. Alzheimer's Disease: Models and Molecular Mechanisms Informing Disease and Treatments. Bioengineering (Basel) 2024; 11:45. [PMID: 38247923 PMCID: PMC10813760 DOI: 10.3390/bioengineering11010045] [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: 11/14/2023] [Revised: 12/15/2023] [Accepted: 12/22/2023] [Indexed: 01/23/2024] Open
Abstract
Alzheimer's Disease (AD) is a complex neurodegenerative disease resulting in progressive loss of memory, language and motor abilities caused by cortical and hippocampal degeneration. This review captures the landscape of understanding of AD pathology, diagnostics, and current therapies. Two major mechanisms direct AD pathology: (1) accumulation of amyloid β (Aβ) plaque and (2) tau-derived neurofibrillary tangles (NFT). The most common variants in the Aβ pathway in APP, PSEN1, and PSEN2 are largely responsible for early-onset AD (EOAD), while MAPT, APOE, TREM2 and ABCA7 have a modifying effect on late-onset AD (LOAD). More recent studies implicate chaperone proteins and Aβ degrading proteins in AD. Several tests, such as cognitive function, brain imaging, and cerebral spinal fluid (CSF) and blood tests, are used for AD diagnosis. Additionally, several biomarkers seem to have a unique AD specific combination of expression and could potentially be used in improved, less invasive diagnostics. In addition to genetic perturbations, environmental influences, such as altered gut microbiome signatures, affect AD. Effective AD treatments have been challenging to develop. Currently, there are several FDA approved drugs (cholinesterase inhibitors, Aß-targeting antibodies and an NMDA antagonist) that could mitigate AD rate of decline and symptoms of distress.
Collapse
Affiliation(s)
- Kaden L. Nystuen
- Department of Chemical Engineering, University of Massachusetts Amherst, Amherst, MA 01003, USA
| | - Shannon M. McNamee
- Schepens Eye Research Institute, Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA 02114, USA
| | - Monica Akula
- Schepens Eye Research Institute, Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA 02114, USA
| | - Kristina M. Holton
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA
- Harvard Stem Cell Institute, Cambridge, MA 02138, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Margaret M. DeAngelis
- Department of Ophthalmology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY 14203, USA
| | - Neena B. Haider
- Schepens Eye Research Institute, Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA 02114, USA
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
10
|
Arendash GW, Lin X, Cao C. Enhanced Brain Clearance of Tau and Amyloid-β in Alzheimer's Disease Patients by Transcranial Radiofrequency Wave Treatment: A Central Role of Vascular Endothelial Growth Factor (VEGF). J Alzheimers Dis 2024; 100:S223-S241. [PMID: 39177605 PMCID: PMC11380279 DOI: 10.3233/jad-240600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/14/2024] [Indexed: 08/24/2024]
Abstract
Background While drainage/removal of fluid and toxins from the brain by cerebrospinal fluid (CSF) directly into venous blood is well-known, a second drainage route has recently been (re)discovered-meningeal lymphatic vessels (mLVs)-which are responsible for up to half of total brain fluid/toxin drainage. The cytokine vascular endothelial growth factor (VEGF) increases mLV diameter and numbers to increase mLV drainage, resulting in increased mLV drainage. Alzheimer's disease (AD) is characterized by low plasma and CSF levels of VEGF. Objective To determine if non-invasive transcranial radiofrequency wave treatment (TRFT), through modulation of VEGF levels in blood and CSF, can affect removal of toxins tau and amyloid-β (Aβ) from the brain. Methods Eight mild/moderate AD subjects were given twice-daily 1-hour TRFT sessions at home by their caregivers. Blood and CSF samples were taken at baseline and following completion of 2 months of TRFT. Results In plasma and/or CSF, strong baseline correlations between VEGF levels and AD markers (t-tau, p-tau, Aβ1-40, Aβ1-42) were eliminated by TRFT. This effect was primarily due to TRFT-induced increases in VEGF levels in AD subjects with low or unmeasurable "baseline" VEGF levels. These increased VEGF levels were associated with increased clearance/drainage of tau and Aβ from the brain, likely through VEGF's actions on mLVs. Conclusions A new mechanism of TRFT is identified (facilitation of brain tau and Aβ clearance via VEGF) that is likely contributory to TRFT's reversal of cognitive impairment in AD subjects. TRFT may be particularly effective for cognitive benefit in AD subjects who have low VEGF levels.
Collapse
Affiliation(s)
- Gary W. Arendash
- RF Longevity, Phoenix, AZ, USA
- NeuroEM Therapeutics, Inc., Tampa, FL, USA
| | - Xiaoyang Lin
- Taneja College of Pharmacy, University of South Florida, Tampa FL, USA
| | - Chuanhai Cao
- Taneja College of Pharmacy, University of South Florida, Tampa FL, USA
- MegaNano Biotech, Tampa, FL, USA
| |
Collapse
|
11
|
Son Y, Park HJ, Jeong YJ, Choi HD, Kim N, Lee HJ. Long-term radiofrequency electromagnetic fields exposure attenuates cognitive dysfunction in 5×FAD mice by regulating microglial function. Neural Regen Res 2023; 18:2497-2503. [PMID: 37282482 DOI: 10.4103/1673-5374.371379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/08/2023] Open
Abstract
We have previously found that long-term effects of exposure to radiofrequency electromagnetic fields in 5×FAD mice with severe late-stage Alzheimer's disease reduced both amyloid-β deposition and glial activation, including microglia. To examine whether this therapeutic effect is due to the regulation of activated microglia, we analyzed microglial gene expression profiles and the existence of microglia in the brain in this study. 5×FAD mice at the age of 1.5 months were assigned to sham- and radiofrequency electromagnetic fields-exposed groups and then animals were exposed to 1950 MHz radiofrequency electromagnetic fields at a specific absorption rate of 5 W/kg for 2 hours/day and 5 days/week for 6 months. We conducted behavioral tests including the object recognition and Y-maze tests and molecular and histopathological analysis of amyloid precursor protein/amyloid-beta metabolism in brain tissue. We confirmed that radiofrequency electromagnetic field exposure for 6 months ameliorated cognitive impairment and amyloid-β deposition. The expression levels of Iba1 (pan-microglial marker) and colony-stimulating factor 1 receptor (CSF1R; regulates microglial proliferation) in the hippocampus in 5×FAD mice treated with radiofrequency electromagnetic fields were significantly reduced compared with those of the sham-exposed group. Subsequently, we analyzed the expression levels of genes related to microgliosis and microglial function in the radiofrequency electromagnetic fields-exposed group compared to those of a CSF1R inhibitor (PLX3397)-treated group. Both radiofrequency electromagnetic fields and PLX3397 suppressed the levels of genes related to microgliosis (Csf1r, CD68, and Ccl6) and pro-inflammatory cytokine interleukin-1β. Notably, the expression levels of genes related to microglial function, including Trem2, Fcgr1a, Ctss, and Spi1, were decreased after long-term radiofrequency electromagnetic field exposure, which was also observed in response to microglial suppression by PLX3397. These results showed that radiofrequency electromagnetic fields ameliorated amyloid-β pathology and cognitive impairment by suppressing amyloid-β deposition-induced microgliosis and their key regulator, CSF1R.
Collapse
Affiliation(s)
- Yeonghoon Son
- Division of Radiation Biomedical Research, Korea Institute of Radiological & Medical Sciences, Seoul, Korea
| | - Hye-Jin Park
- Division of Radiation Biomedical Research, Korea Institute of Radiological & Medical Sciences, Seoul, Korea
| | - Ye Ji Jeong
- Division of Radiation Biomedical Research, Korea Institute of Radiological & Medical Sciences, Seoul, Korea
| | - Hyung-Do Choi
- Department of EMF Research Team, Radio and Broadcasting Technology Laboratory, Electronics and Telecommunications Research Institute, Daejon, Korea
| | - Nam Kim
- School of Electrical and Computer Engineering, Chungbuk National University, Cheongju, Korea
| | - Hae-June Lee
- Division of Radiation Biomedical Research, Korea Institute of Radiological & Medical Sciences, Seoul, Korea
| |
Collapse
|
12
|
El-Kafoury BMA, Abdel-Hady EA, El Bakly W, Elayat WM, Hamam GG, Abd El Rahman SMM, Lasheen NN. Lipoic acid inhibits cognitive impairment induced by multiple cell phones in young male rats: role of Sirt1 and Atg7 pathway. Sci Rep 2023; 13:18486. [PMID: 37898621 PMCID: PMC10613255 DOI: 10.1038/s41598-023-44134-2] [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: 03/11/2023] [Accepted: 10/04/2023] [Indexed: 10/30/2023] Open
Abstract
The utilization of digital technology has grown rapidly in the past three decades. With this rapid increase, cell phones emit electromagnetic radiation; that is why electromagnetic field (EMF) has become a substantial new pollution source in modern civilization, mainly having adverse effects on the brain. While such a topic attracted many researchers' scopes, there are still minimal discoveries made regarding chronic exposure to EMF. The extensive use of cell phones may affect children's cognition even indirectly if parents and guardians used their phones repeatedly near them. This study aims to investigate possible lipoic acid (LA) effects on cognitive functions and hippocampal structure in young male rats exposed to electromagnetic fields (EMF) emitted from multiple cell phones. Forty young male Wistar rats were randomly allocated into three groups: control, multiple cell phones-exposed and lipoic acid-treated rats. By the end of the experimental period, the Morris water maze was used as a cognitive test. The rats were sacrificed for the collection of serum and hippocampal tissue. These serum samples were then utilized for assessment of Liver function tests. The level ofglutamate, acetylcholine (Ach) and malondialdehyde (MDA) was estimated, in addition to evaluating the expression of autophagy-related protein-7 (Atg7) and Sirt1 genes. The left hippocampal specimens were used for histopathological studies. Results showed that multiple cell phone-exposed rats exhibited shorter latency time to reach the platform by the fifth day of training; additionally, there was a reduction in consolidation of spatial long-term memory. Correspondingly, there was an elevation of hippocampal Ach, glutamate, and MDA levels; accompanied by up-regulation of hippocampal Sirt1 and Atg7 gene expression. Compared to the EMF-exposed group, LA administration improved both learning and memory, this was proved by the significant decline in hippocampal MDA and Ach levels, the higher hippocampal glutamate, the downregulated hippocampal Sirt1 gene expression and the upregulated Atg7 gene expression. In conclusion, EMF exposure could enhance learning ability; however, it interfered with long-term memory consolidation shown by higher hippocampal Ach levels. Lipoic acid treatment improved both learning and memory by enhancing autophagy and hippocampal glutamate level and by the reduced Ach levels and Sirt1 gene expression.
Collapse
Affiliation(s)
- Bataa M A El-Kafoury
- Department of Medical Physiology, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Enas A Abdel-Hady
- Department of Medical Physiology, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Wesam El Bakly
- Department of Clinical Pharmacology, Faculty of Medicine, Ain Shams University, Cairo, Egypt
- Department of Medical Pharmacology, Faculty of Medicine, AFCM, Cairo, Egypt
| | - Wael M Elayat
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, Ain Shams University, Cairo, Egypt
- Department of Basic Medical Sciences, Faculty of Medicine, Galala University, Galala City, Egypt
| | - Ghada Galal Hamam
- Department of Histology and Cell Biology, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | | | - Noha N Lasheen
- Department of Medical Physiology, Faculty of Medicine, Ain Shams University, Cairo, Egypt.
- Department of Basic Medical Sciences, Faculty of Medicine, Galala University, Galala City, Egypt.
| |
Collapse
|
13
|
Arendash G, Cao C. Transcranial Electromagnetic Wave Treatment: A Fountain of Healthy Longevity? Int J Mol Sci 2023; 24:ijms24119652. [PMID: 37298603 DOI: 10.3390/ijms24119652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 05/29/2023] [Accepted: 05/30/2023] [Indexed: 06/12/2023] Open
Abstract
Most diseases of older age have as their common denominator a dysfunctional immune system, wherein a low, chronic level of inflammation is present due to an imbalance of pro-inflammatory cytokines over anti-inflammatory cytokines that develops during aging ("inflamm-aging"). A gerotherapeutic that can restore the immune balance to that shared by young/middle-aged adults and many centenarians could reduce the risk of those age-related diseases and increase healthy longevity. In this perspectives paper, we discuss potential longevity interventions that are being evaluated and compare them to a novel gerotherapeutic currently being evaluated in humans-Transcranial Electromagnetic Wave Treatment (TEMT). TEMT is provided non-invasively and safety through a novel bioengineered medical device-the MemorEM-that allows for near complete mobility during in-home treatments. Daily TEMT to mild/moderate Alzheimer's Disease (AD) patients over a 2-month period rebalanced 11 of 12 cytokines in blood back to that of normal aged adults. A very similar TEMT-induced rebalancing of cytokines occurred in the CSF/brain for essentially all seven measurable cytokines. Overall inflammation in both blood and brain was dramatically reduced by TEMT over a 14-27 month period, as measured by C-Reactive Protein. In these same AD patients, a reversal of cognitive impairment was observed at 2 months into treatment, while cognitive decline was stopped over a 2½ year period of TEMT. Since most age-related diseases have the commonality of immune imbalance, it is reasonable to postulate that TEMT could rebalance the immune system in many age-related diseases as it appears to do in AD. We propose that TEMT has the potential to reduce the risk/severity of age-related diseases by rejuvenating the immune system to a younger age, resulting in reduced brain/body inflammation and a substantial increase in healthy longevity.
Collapse
Affiliation(s)
- Gary Arendash
- NeuroEM Therapeutics, Inc., 501 E. Kennedy Blvd., Suite 650, Tampa, FL 33602, USA
| | - Chuanhai Cao
- Taneja College of Pharmacy, University of South Florida, Tampa, FL 33612, USA
- MegaNano Biotech, 3802 Spectrum Blvd., Suite 122, Tampa, FL 33612, USA
| |
Collapse
|
14
|
Zheng R, Zhang X, Gao Y, Gao D, Gong W, Zhang C, Dong G, Li Z. Biological effects of exposure to 2650 MHz electromagnetic radiation on the behavior, learning, and memory of mice. Brain Behav 2023; 13:e3004. [PMID: 37118929 PMCID: PMC10275548 DOI: 10.1002/brb3.3004] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 03/09/2023] [Accepted: 03/27/2023] [Indexed: 04/30/2023] Open
Abstract
BACKGROUND With the development of communication technology, the public is paying increasing attention to whether electromagnetic radiation is harmful to health. Mobile phone communication has entered the 5G era, and there are almost no reports on electromagnetic radiation at 2650 MHz. Therefore, it is necessary to evaluate the risk of adverse effects of 5G mobile phone EMR exposure on the human brain. METHODS Male animals were continuously exposed to 2650 MHz-EMR for 28 days with a whole-body averaged specific absorption rate (WBSAR) of 2.06 W/kg for 4 h per day. Mouse behavior was assessed using the open-field test (OFT), elevated-plus maze (EPM), and tail suspension test (TST). The Morris water maze (MWM), HE staining, and TUNEL staining were used to evaluate the spatial memory ability and pathological morphology of hippocampal dentate gyrus cells. Additionally, the expression levels of brain-derived neurotrophic factor (BDNF), aminobutyric acid (GABA), and glucocorticoid (GR) in the hippocampus were detected by western blotting and immunohistochemistry, while the corticosterone (CORT) level in serum was detected by ELISA. RESULTS In the OFT, the total distance traveled, central distance traveled, and residence time significantly decreased in the EMR exposure group (p < .05). In EPM, the percentage of the number of times to open the arm and the percentage of time to open the arm significantly decreased in the EMR exposure group. However, in the TST, the two groups had no significant difference in the 4-min immobility time. In the MWM, the escape latency of the EMR exposure group was shorter than that of the control group, with no significant difference. Furthermore, CORT levels in serum were significantly increased in the EMR exposure group (p < .05), while the expression of BDNF and GR proteins in the hippocampus was reduced (p < .05), but there was no significant difference in GABA expression. CONCLUSIONS Our results indicate that exposure to 2650 MHz-EMR (WBSAR: 2.06 W/kg, 28 days, 4 h per day) had no significant effect on the spatial memory ability of mice (in comparison to little effect). The exposure may be associated with anxiety-like behavior in mice but not related to depression-like behavior in mice.
Collapse
Affiliation(s)
- Rongqi Zheng
- Laboratory of Electromagnetic Biological Effects, Beijing Insititute of Radiation and MedicineBeijingChina
| | - Xianxie Zhang
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation and MedicineBeijingChina
| | - Yan Gao
- Laboratory of Electromagnetic Biological Effects, Beijing Insititute of Radiation and MedicineBeijingChina
| | - Dawen Gao
- Laboratory of Electromagnetic Biological Effects, Beijing Insititute of Radiation and MedicineBeijingChina
| | - Wenjing Gong
- Laboratory of Electromagnetic Biological Effects, Beijing Insititute of Radiation and MedicineBeijingChina
| | - Chenggang Zhang
- Laboratory of Electromagnetic Biological Effects, Beijing Insititute of Radiation and MedicineBeijingChina
| | - Guofu Dong
- Laboratory of Electromagnetic Biological Effects, Beijing Insititute of Radiation and MedicineBeijingChina
| | - Zhihui Li
- Laboratory of Electromagnetic Biological Effects, Beijing Insititute of Radiation and MedicineBeijingChina
| |
Collapse
|
15
|
Zhi W, Yong Z, Ma L, He S, Guo Z, Zhao X, Hu X, Wang L. 900 MHz electromagnetic field exposure relieved AD-like symptoms on APP/PS1 mice: A potential non-invasive strategy for AD treatment. Biochem Biophys Res Commun 2023; 658:97-106. [PMID: 37030070 DOI: 10.1016/j.bbrc.2023.03.083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 03/15/2023] [Accepted: 03/31/2023] [Indexed: 04/04/2023]
Abstract
BACKGROUND Evidence shows that microwaves radiation may have various biological effects on central nervous system. Role of electromagnetic fields in neurodegenerative diseases, especially AD, has been widely studied, but results of these studies are inconsistent. Therefore, the above effects were verified again and the mechanism was preliminarily discussed. METHODS Amyloid precursor protein (APP/PS1) and WT mice were exposed to long-term microwave radiation for 270 days (900 MHz, SAR: 0.25-1.055 W/kg, 2 h/day, alternately), and related indices were assessed at 90, 180 and 270 days. Cognition was evaluated by Morris water maze, Y maze and new object recognition tests. Congo red staining, immunohistochemistry and ELISA were used to analyze Aβ plaques, Aβ40 and Aβ42 content. Differentially expressed proteins in hippocampus between microwave-exposed and unexposed AD mice were identified by proteomics. RESULTS Spatial and working memory was improved in AD mice after long-term 900 MHz microwave exposure compared with after sham exposure. Microwave radiation (900 MHz) for 180 or 270 days did not induce Aβ plaque formation in WT mice but inhibited Aβ accumulation in the cerebral cortex and hippocampus in 2- and 5-month-old APP/PS1 mice. This effect mainly occurred in the late stage of the disease and may have been attributed to downregulation of apolipoprotein family member and SNCA expression and excitatory/inhibitory neurotransmitter rebalance in the hippocampus. CONCLUSIONS The present results indicated that long-term microwave radiation can retard AD development and exert a beneficial effect against AD, suggesting that 900 MHz microwave exposure may be a potential therapy for AD.
Collapse
|
16
|
Bayat M, Karimi N, Karami M, Haghighi AB, Bayat K, Akbari S, Haghani M. Chronic exposure to 2.45 GHz microwave radiation improves cognition and synaptic plasticity impairment in vascular dementia model. Int J Neurosci 2023; 133:111-122. [PMID: 33635159 DOI: 10.1080/00207454.2021.1896502] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Purpose: In this study, we evaluated the effects of 2.45 GHz microwave radiation on cognitive dysfunction induced by vascular dementia (VaD).Methods: The VaD was induced by bilateral-common carotid occlusion (2-VO). The rats were divided into 4 groups including: control (n = 6), sham (n = 6), 2-VO (n = 8), and 2-VO + Wi-Fi (n = 10) groups. Wi-Fi modem centrally located at the distance of 25 cm from the animal's cages and the animals were continuously exposed to Wi-Fi signal while they freely moved in the cage (2 h/day for forty-five days). Therefore, the power density (PD) and specific absorption rate value (SAR) decreased at a distance of 25 to 60 cm (PD = 0.018 to 0.0032 mW/cm2, SAR = 0.0346 to 0.0060 W/Kg). The learning, memory, and hippocampal synaptic-plasticity were evaluated by radial arm maze (RAM), passive avoidance (PA), and field-potential recording respectively. The number of hippocampal CA1 cells was also assessed by giemsa staining.Results: Our results showed that VaD model led to impairment in the spatial learning and memory performance in RAM and PA that were associated with long-term potentiation (LTP) impairment, decrease of basal-synaptic transmission (BST), increase of GABA transmission, and decline of neurotransmitter release-probability as well as hippocampal cell loss. Notably, chronic Wi-Fi exposure significantly recovered the learning-memory performance, LTP induction, and cell loss without any effect on BST.Conclusions: The LTP recovery by Wi-Fi in the 2-VO rats was probably related to significant increases in the hippocampal CA1 neuronal density, partial recovery of neurotransmitter release probability, and reduction of GABA transmissiSon as evident by rescue of paired-pulse ratio 10 ms.
Collapse
Affiliation(s)
- Mahnaz Bayat
- Clinical Neurology Research Centre, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Narges Karimi
- Department of Physiology, the Medical School, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohammad Karami
- Department of Physiology, the Medical School, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | - Kamjoo Bayat
- Department of Physics, K. N. Toosi University of Technology, Tehran, Iran
| | - Somayeh Akbari
- Department of Physiology, the Medical School, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Masoud Haghani
- Department of Physiology, the Medical School, Shiraz University of Medical Sciences, Shiraz, Iran.,Histomorphometry and Stereology Research Centre, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
17
|
Abkhezr H, Mohaddes G, Nikniaz Z, Abbasalizad Farhangi M, Heydari H, Nikniaz L. The effect of Extremely Low Frequency Electromagnetic Field on spatial memory of mice and rats: A systematic review. LEARNING AND MOTIVATION 2023. [DOI: 10.1016/j.lmot.2023.101873] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/13/2023]
|
18
|
Transcranial Electromagnetic Treatment Stops Alzheimer’s Disease Cognitive Decline over a 2½-Year Period: A Pilot Study. MEDICINES 2022; 9:medicines9080042. [PMID: 36005647 PMCID: PMC9416517 DOI: 10.3390/medicines9080042] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 06/29/2022] [Accepted: 07/13/2022] [Indexed: 11/16/2022]
Abstract
Background: There is currently no therapeutic that can stop or reverse the progressive memory impairment of Alzheimer’s disease (AD). However, we recently published that 2 months of daily, in-home transcranial electromagnetic treatment (TEMT) reversed the cognitive impairment in eight mild/moderate AD subjects. These cognitive enhancements were accompanied by predicted changes in AD markers within both the blood and cerebrospinal fluid (CSF). Methods: In view of these encouraging findings, the initial clinical study was extended twice to encompass a period of 2½ years. The present study reports on the resulting long-term safety, cognitive assessments, and AD marker evaluations from the five subjects who received long-term treatment. Results: TEMT administration was completely safe over the 2½-year period, with no deleterious side effects. In six cognitive/functional tasks (including the ADAS-cog13, Rey AVLT, MMSE, and ADL), no decline in any measure occurred over this 2½-year period. Long-term TEMT induced reductions in the CSF levels of C-reactive protein, p-tau217, Aβ1-40, and Aβ1-42 while modulating CSF oligomeric Aβ levels. In the plasma, long-term TEMT modulated/rebalanced levels of both p-tau217 and total tau. Conclusions: Although only a limited number of AD patients were involved in this study, the results suggest that TEMT can stop the cognitive decline of AD over a period of at least 2½ years and can do so with no safety issues.
Collapse
|
19
|
Perez FP, Bandeira JP, Perez Chumbiauca CN, Lahiri DK, Morisaki J, Rizkalla M. Multidimensional insights into the repeated electromagnetic field stimulation and biosystems interaction in aging and age-related diseases. J Biomed Sci 2022; 29:39. [PMID: 35698225 PMCID: PMC9190166 DOI: 10.1186/s12929-022-00825-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 06/07/2022] [Indexed: 11/25/2022] Open
Abstract
We provide a multidimensional sequence of events that describe the electromagnetic field (EMF) stimulation and biological system interaction. We describe this process from the quantum to the molecular, cellular, and organismal levels. We hypothesized that the sequence of events of these interactions starts with the oscillatory effect of the repeated electromagnetic stimulation (REMFS). These oscillations affect the interfacial water of an RNA causing changes at the quantum and molecular levels that release protons by quantum tunneling. Then protonation of RNA produces conformational changes that allow it to bind and activate Heat Shock Transcription Factor 1 (HSF1). Activated HSF1 binds to the DNA expressing chaperones that help regulate autophagy and degradation of abnormal proteins. This action helps to prevent and treat diseases such as Alzheimer's and Parkinson's disease (PD) by increasing clearance of pathologic proteins. This framework is based on multiple mathematical models, computer simulations, biophysical experiments, and cellular and animal studies. Results of the literature review and our research point towards the capacity of REMFS to manipulate various networks altered in aging (Reale et al. PloS one 9, e104973, 2014), including delay of cellular senescence (Perez et al. 2008, Exp Gerontol 43, 307-316) and reduction in levels of amyloid-β peptides (Aβ) (Perez et al. 2021, Sci Rep 11, 621). Results of these experiments using REMFS at low frequencies can be applied to the treatment of patients with age-related diseases. The use of EMF as a non-invasive therapeutic modality for Alzheimer's disease, specifically, holds promise. It is also necessary to consider the complicated and interconnected genetic and epigenetic effects of the REMFS-biological system's interaction while avoiding any possible adverse effects.
Collapse
Affiliation(s)
- Felipe P Perez
- Indiana University School of Medicine, Indianapolis, IN, USA.
- Division of General Internal Medicine and Geriatrics, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA.
| | - Joseph P Bandeira
- Indiana University School of Medicine, Indianapolis, IN, USA
- Division of General Internal Medicine and Geriatrics, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Cristina N Perez Chumbiauca
- Indiana University School of Medicine, Indianapolis, IN, USA
- Division of Rheumatology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Debomoy K Lahiri
- Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Psychiatry, Institute of Psychiatric Research, Neuroscience Research Center, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Jorge Morisaki
- Department of Bioengineering, University of Illinois at Chicago, Chicago, IL, USA
| | - Maher Rizkalla
- Department of Electrical and Computer Engineering, Indiana University-Purdue University, Indianapolis, IN, USA
| |
Collapse
|
20
|
Cao C, Abulaban H, Baranowski R, Wang Y, Bai Y, Lin X, Shen N, Zhang X, Arendash GW. Transcranial Electromagnetic Treatment “Rebalances” Blood and Brain Cytokine Levels in Alzheimer’s Patients: A New Mechanism for Reversal of Their Cognitive Impairment. Front Aging Neurosci 2022; 14:829049. [PMID: 35585867 PMCID: PMC9108275 DOI: 10.3389/fnagi.2022.829049] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2021] [Accepted: 04/05/2022] [Indexed: 01/11/2023] Open
Abstract
Background The immune system plays a critical role in the development and progression of Alzheimer’s disease (AD). However, there is disagreement as to whether development/progression of AD involves an over-activation or an under-activation of the immune system. In either scenario, the immune system’s cytokine levels are abnormal in AD and in need of rebalancing. We have recently published a pilot clinical trial (https://clinicaltrials.gov/ct2/show/NCT02958930) showing that 2 months of daily in-home Transcranial Electromagnetic Treatment (TEMT) was completely safe and resulted in reversal of AD cognitive impairment. Methods For the eight mild/moderate AD subjects in this published work, the present study sought to determine if their TEMT administration had immunologic effects on blood or CSF levels of 12 cytokines. Subjects were given daily in-home TEMT for 2 months by their caregivers, utilizing first-in-class MemorEM™ devices. Results For eight plasma cytokines, AD subjects with lower baseline cytokine levels always showed increases in those cytokines after both a single treatment or after 2-months of daily TEMT. By contrast, those AD subjects with higher baseline cytokine levels in plasma showed treatment-induced decreases in plasma cytokines at both time points. Thus, a gravitation to reported normal plasma cytokine levels (i.e., a “rebalancing”) occurred with both acute and long-term TEMT. In the CSF, TEMT-induced a similar rebalancing for seven measurable cytokines, the direction and extent of changes in individual subjects also being linked to their baseline CSF levels. Conclusion Our results strongly suggest that daily TEMT to AD subjects for 2-months can “rebalance” levels for 11 of 12 cytokines in blood and/or brain, which is associated with reversal of their cognitive impairment. TEMT is likely to be providing these immunoregulatory effects by affecting cytokine secretion from: (1) blood cells traveling through the head’s vasculature, and (2) the brain’s microglia/astrocytes, choroid plexus, or neurons. This rebalancing of so many cytokines, and in both brain and systemic compartments, appears to be a remarkable new mechanism of TEMT action that may contribute substantially to it’s potential to prevent, stop, or reverse AD and other diseases of aging.
Collapse
Affiliation(s)
- Chuanhai Cao
- Taneja College of Pharmacy, University of South Florida, Tampa, FL, United States
- MegaNano Biotech, Inc., Tampa, FL, United States
| | - Haitham Abulaban
- Axiom Clinical Research, Tampa, FL, United States
- University of South Florida Health Byrd Alzheimer’s Institute, Tampa, FL, United States
| | | | - Yanhong Wang
- Taneja College of Pharmacy, University of South Florida, Tampa, FL, United States
| | - Yun Bai
- MegaNano Biotech, Inc., Tampa, FL, United States
| | - Xiaoyang Lin
- Taneja College of Pharmacy, University of South Florida, Tampa, FL, United States
- MegaNano Biotech, Inc., Tampa, FL, United States
| | - Ning Shen
- Taneja College of Pharmacy, University of South Florida, Tampa, FL, United States
- MegaNano Biotech, Inc., Tampa, FL, United States
| | - Xiaolin Zhang
- Taneja College of Pharmacy, University of South Florida, Tampa, FL, United States
- MegaNano Biotech, Inc., Tampa, FL, United States
| | - Gary W. Arendash
- NeuroEM Therapeutics, Inc., Phoenix, AZ, United States
- *Correspondence: Gary W. Arendash,
| |
Collapse
|
21
|
Nguyen PH, Ramamoorthy A, Sahoo BR, Zheng J, Faller P, Straub JE, Dominguez L, Shea JE, Dokholyan NV, De Simone A, Ma B, Nussinov R, Najafi S, Ngo ST, Loquet A, Chiricotto M, Ganguly P, McCarty J, Li MS, Hall C, Wang Y, Miller Y, Melchionna S, Habenstein B, Timr S, Chen J, Hnath B, Strodel B, Kayed R, Lesné S, Wei G, Sterpone F, Doig AJ, Derreumaux P. Amyloid Oligomers: A Joint Experimental/Computational Perspective on Alzheimer's Disease, Parkinson's Disease, Type II Diabetes, and Amyotrophic Lateral Sclerosis. Chem Rev 2021; 121:2545-2647. [PMID: 33543942 PMCID: PMC8836097 DOI: 10.1021/acs.chemrev.0c01122] [Citation(s) in RCA: 455] [Impact Index Per Article: 113.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Protein misfolding and aggregation is observed in many amyloidogenic diseases affecting either the central nervous system or a variety of peripheral tissues. Structural and dynamic characterization of all species along the pathways from monomers to fibrils is challenging by experimental and computational means because they involve intrinsically disordered proteins in most diseases. Yet understanding how amyloid species become toxic is the challenge in developing a treatment for these diseases. Here we review what computer, in vitro, in vivo, and pharmacological experiments tell us about the accumulation and deposition of the oligomers of the (Aβ, tau), α-synuclein, IAPP, and superoxide dismutase 1 proteins, which have been the mainstream concept underlying Alzheimer's disease (AD), Parkinson's disease (PD), type II diabetes (T2D), and amyotrophic lateral sclerosis (ALS) research, respectively, for many years.
Collapse
Affiliation(s)
- Phuong H Nguyen
- CNRS, UPR9080, Université de Paris, Laboratory of Theoretical Biochemistry, IBPC, Fondation Edmond de Rothschild, PSL Research University, Paris 75005, France
| | - Ayyalusamy Ramamoorthy
- Biophysics and Department of Chemistry, University of Michigan, Ann Arbor, Michigan 48109-1055, United States
| | - Bikash R Sahoo
- Biophysics and Department of Chemistry, University of Michigan, Ann Arbor, Michigan 48109-1055, United States
| | - Jie Zheng
- Department of Chemical & Biomolecular Engineering, The University of Akron, Akron, Ohio 44325, United States
| | - Peter Faller
- Institut de Chimie, UMR 7177, CNRS-Université de Strasbourg, 4 rue Blaise Pascal, 67000 Strasbourg, France
| | - John E Straub
- Department of Chemistry, Boston University, 590 Commonwealth Avenue, Boston, Massachusetts 02215, United States
| | - Laura Dominguez
- Facultad de Química, Departamento de Fisicoquímica, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico
| | - Joan-Emma Shea
- Department of Chemistry and Biochemistry, and Department of Physics, University of California, Santa Barbara, California 93106, United States
| | - Nikolay V Dokholyan
- Department of Pharmacology and Biochemistry & Molecular Biology, Penn State University College of Medicine, Hershey, Pennsylvania 17033, United States
- Department of Chemistry, and Biomedical Engineering, Pennsylvania State University, University Park, Pennsylvania 16802, United States
| | - Alfonso De Simone
- Department of Life Sciences, Imperial College London, London SW7 2AZ, U.K
- Molecular Biology, University of Naples Federico II, Naples 80138, Italy
| | - Buyong Ma
- Basic Science Program, Leidos Biomedical Research, Inc., Cancer and Inflammation Program, National Cancer Institute, Frederick, Maryland 21702, United States
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai, China
| | - Ruth Nussinov
- Basic Science Program, Leidos Biomedical Research, Inc., Cancer and Inflammation Program, National Cancer Institute, Frederick, Maryland 21702, United States
- Sackler Institute of Molecular Medicine, Department of Human Genetics and Molecular Medicine Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Saeed Najafi
- Department of Chemistry and Biochemistry, and Department of Physics, University of California, Santa Barbara, California 93106, United States
| | - Son Tung Ngo
- Laboratory of Theoretical and Computational Biophysics & Faculty of Applied Sciences, Ton Duc Thang University, 33000 Ho Chi Minh City, Vietnam
| | - Antoine Loquet
- Institute of Chemistry & Biology of Membranes & Nanoobjects, (UMR5248 CBMN), CNRS, Université Bordeaux, Institut Européen de Chimie et Biologie, 33600 Pessac, France
| | - Mara Chiricotto
- Department of Chemical Engineering and Analytical Science, University of Manchester, Manchester M13 9PL, U.K
| | - Pritam Ganguly
- Department of Chemistry and Biochemistry, and Department of Physics, University of California, Santa Barbara, California 93106, United States
| | - James McCarty
- Chemistry Department, Western Washington University, Bellingham, Washington 98225, United States
| | - Mai Suan Li
- Institute for Computational Science and Technology, SBI Building, Quang Trung Software City, Tan Chanh Hiep Ward, District 12, Ho Chi Minh City 700000, Vietnam
- Institute of Physics, Polish Academy of Sciences, Al. Lotnikow 32/46, 02-668 Warsaw, Poland
| | - Carol Hall
- Department of Chemical and Biomolecular Engineering, North Carolina State University, Raleigh, North Carolina 27695-7905, United States
| | - Yiming Wang
- Department of Chemical and Biomolecular Engineering, North Carolina State University, Raleigh, North Carolina 27695-7905, United States
| | - Yifat Miller
- Department of Chemistry and The Ilse Katz Institute for Nanoscale Science & Technology, Ben-Gurion University of the Negev, Be'er Sheva 84105, Israel
| | | | - Birgit Habenstein
- Institute of Chemistry & Biology of Membranes & Nanoobjects, (UMR5248 CBMN), CNRS, Université Bordeaux, Institut Européen de Chimie et Biologie, 33600 Pessac, France
| | - Stepan Timr
- CNRS, UPR9080, Université de Paris, Laboratory of Theoretical Biochemistry, IBPC, Fondation Edmond de Rothschild, PSL Research University, Paris 75005, France
| | - Jiaxing Chen
- Department of Pharmacology and Biochemistry & Molecular Biology, Penn State University College of Medicine, Hershey, Pennsylvania 17033, United States
| | - Brianna Hnath
- Department of Pharmacology and Biochemistry & Molecular Biology, Penn State University College of Medicine, Hershey, Pennsylvania 17033, United States
| | - Birgit Strodel
- Institute of Complex Systems: Structural Biochemistry (ICS-6), Forschungszentrum Jülich, 52425 Jülich, Germany
| | - Rakez Kayed
- Mitchell Center for Neurodegenerative Diseases, and Departments of Neurology, Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, Texas 77555, United States
| | - Sylvain Lesné
- Department of Neuroscience, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Guanghong Wei
- Department of Physics, State Key Laboratory of Surface Physics, and Key Laboratory for Computational Physical Science, Multiscale Research Institute of Complex Systems, Fudan University, Shanghai 200438, China
| | - Fabio Sterpone
- CNRS, UPR9080, Université de Paris, Laboratory of Theoretical Biochemistry, IBPC, Fondation Edmond de Rothschild, PSL Research University, Paris 75005, France
| | - Andrew J Doig
- Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PT, U.K
| | - Philippe Derreumaux
- CNRS, UPR9080, Université de Paris, Laboratory of Theoretical Biochemistry, IBPC, Fondation Edmond de Rothschild, PSL Research University, Paris 75005, France
- Laboratory of Theoretical Chemistry, Ton Duc Thang University, 33000 Ho Chi Minh City, Vietnam
- Faculty of Pharmacy, Ton Duc Thang University, 33000 Ho Chi Minh City, Vietnam
| |
Collapse
|
22
|
Ansari AH, Pal A, Ramamurthy A, Kabat M, Jain S, Kumar S. Fibromyalgia Pain and Depression: An Update on the Role of Repetitive Transcranial Magnetic Stimulation. ACS Chem Neurosci 2021; 12:256-270. [PMID: 33397091 DOI: 10.1021/acschemneuro.0c00785] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Fibromyalgia is a musculoskeletal pain of different parts of the body, which is also associated with fatigue, lack of sleep, cognition deficits, family history, gender bias, and other disorders such as osteoarthritis and rheumatoid arthritis. It is generally initiated after trauma, surgery, infection, or stress. Fibromyalgia often coexists with several other conditions or disorders such as temporomandibular joint disorders, bowel and bladder syndrome, anxiety, depression, headaches, and interstitial cystitis. While there is no permanent cure for fibromyalgia, some interventions are available with multiple side effects. rTMS (repetitive transcranial magnetic stimulation), a noninvasive management strategy is used widely for various pain-related etiologies including fibromyalgia in both the laboratory and clinical settings. In this Review, we discuss the role and mechanism of action of rTMS in fibromyalgia patients and on associated comorbidities including anxiety, pain, depression, neurotransmitter alterations, sleep disorders, and overall quality of life of the patients suffering from this chronic problem. We also provide an update on the rTMS application in the clinical trials of fibromyalgia patients and prospective management therapy for multiple problems that these patients suffer.
Collapse
Affiliation(s)
- Abdul Haque Ansari
- Department of Physiology, College of Medicine, Texila American University, East Bank, Demerara, Guyana, South America
| | - Ajay Pal
- Department of Orthopedic Surgery, Movement Recovery Laboratory, Columbia University Medical Center, New York, New York 10032, United States
| | - Aditya Ramamurthy
- Department of Orthopedic Surgery, Movement Recovery Laboratory, Columbia University Medical Center, New York, New York 10032, United States
| | - Maciej Kabat
- Hackensack Meridian School of Medicine, Seton Hall University Interprofessional Health Sciences Campus, Kingsland Street, Nutley, New Jersey 07110, United States
| | - Suman Jain
- Department of Physiology, All India Institute of Medical Sciences, New Delhi 110029, India
| | - Suneel Kumar
- Department of Biomedical Engineering, Rutgers, The State University of New Jersey, Piscataway, New Jersey 08854, United States
| |
Collapse
|
23
|
Maldonado-Moreles A, Cordova-Fraga T, Bonilla-Jaime H, Lopez-Camacho PY, Basurto-Islas G. Low frequency vortex magnetic field reduces amyloid β aggregation, increase cell viability and protect from amyloid β toxicity. Electromagn Biol Med 2021; 40:191-200. [PMID: 33043710 DOI: 10.1080/15368378.2020.1830288] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Accepted: 09/18/2020] [Indexed: 01/05/2023]
Abstract
Plaques formed by abnormal accumulation of amyloid β-peptide (Aβ) lead to onset of Alzheimer's disease (AD). Pharmacological treatments do not reduce Aβ aggregation neither restore learning and memory. Noninvasive techniques have emerged as an alternative to treat AD, such as stimulation with electromagnetic fields (EMF) that decrease Aβ deposition and reverses cognitive impairment in AD mice, even though some studies showed side effects on parallel magnetic fields stimulation. As a new approach of magnetic field (MF) stimulation, vortex magnetic fields (VMF) have been tested inducing a random movement of charged biomolecules in cells, promoting cell viability and apparently safer than parallel magnetic fields. In this study we demonstrate the effect of VMF on Aβ aggregation. The experimental strategy includes, i) design and construction of a coil capable to induce VMF, ii) evaluation of VMF stimulation on Aβ peptide induced-fibrils-formation, iii) evaluation of VMF stimulation on SH-SY5Y neuroblastoma cell line in the presence of Aβ peptide. We demonstrated for the first time that Aβ aggregation exposed to VMF during 24 h decreased ~ 86% of Aβ fibril formation compared to control. Likewise, VMF stimulation reduced Aβ fibrils-cytotoxicity and increase SH-SY5Y cell viability. These data establish the basis for future investigation that involve VMF as inhibitor of Aβ-pathology and indicate the therapeutic potential of VMF for AD treatment.
Collapse
Affiliation(s)
- Alejandro Maldonado-Moreles
- Doctorado en Ciencias Biologicas y de la Salud, Universidad Autonoma Metropolitana , Ciudad de México, México
| | | | - Herlinda Bonilla-Jaime
- Departamento de Biología de la Reproducción, Lab de Psicobiología, Universidad Autónoma Metropolitana Iztapalapa , Ciudad de México, México
| | - Perla Y Lopez-Camacho
- Departamento de Ciencias Naturales, Universidad Autónoma Metropolitana Cuajimalpa , Ciudad de México, México
| | | |
Collapse
|
24
|
Bouji M, Lecomte A, Gamez C, Blazy K, Villégier AS. Impact of Cerebral Radiofrequency Exposures on Oxidative Stress and Corticosterone in a Rat Model of Alzheimer's Disease. J Alzheimers Dis 2020; 73:467-476. [PMID: 31796670 DOI: 10.3233/jad-190593] [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] [Indexed: 12/13/2022]
Abstract
BACKGROUND Alzheimer's disease (AD) is the most common type of neurodegenerative disease leading to dementia. Several studies suggested that mobile phone radiofrequency electromagnetic field (RF-EMF) exposures modified AD memory deficits in rodent models. OBJECTIVE Here we aimed to test the hypothesis that RF-EMF exposure may modify memory through corticosterone and oxidative stress in the Samaritan rat model of AD. METHODS Long-Evans male rats received intracerebroventricular infusion with ferrous sulphate, amyloid-beta 1-42 peptide, and buthionine-sufloximine (AD rats) or with vehicle (control rats). To mimic cell phone use, RF-EMF were exposed to the head for 1 month (5 days/week, in restraint). To look for hazard thresholds, high brain averaged specific absorption rates (BASAR) were tested: 1.5 W/Kg (15 min), 6 W/Kg (15 min), and 6 W/Kg (45 min). The sham group was in restraint for 45 min. Endpoints were spatial memory in the radial maze, plasmatic corticosterone, heme oxygenase-1 (HO1), and amyloid plaques. RESULTS Results indicated similar corticosterone levels but impaired memory performances and increased cerebral staining of thioflavine and of HO1 in the sham AD rats compared to the controls. A correlative increase of cortical HO1 staining was the only effect of RF-EMF in control rats. In AD rats, RF-EMF exposures induced a correlative increase of hippocampal HO1 staining and reduced corticosterone. DISCUSSION According to our data, neither AD nor control rats showed modified memory after RF-EMF exposures. Unlike control rats, AD rats showed higher hippocampal oxidative stress and reduced corticosterone with the higher BASAR. This data suggests more fragility related to neurodegenerative disease toward RF-EMF exposures.
Collapse
Affiliation(s)
- Marc Bouji
- Unité de Toxicologie Expérimentale, Institut National de l'Environnement Industriel et des Risques (INERIS), Parc Technologique ALATA, Verneuil-en-Halatte, France
| | - Anthony Lecomte
- Unité de Toxicologie Expérimentale, Institut National de l'Environnement Industriel et des Risques (INERIS), Parc Technologique ALATA, Verneuil-en-Halatte, France.,PERITOX UMR I-01 INERIS 01 UFR de médecine, Amiens, France
| | - Christelle Gamez
- Unité de Toxicologie Expérimentale, Institut National de l'Environnement Industriel et des Risques (INERIS), Parc Technologique ALATA, Verneuil-en-Halatte, France.,PERITOX UMR I-01 INERIS 01 UFR de médecine, Amiens, France
| | - Kelly Blazy
- Unité de Toxicologie Expérimentale, Institut National de l'Environnement Industriel et des Risques (INERIS), Parc Technologique ALATA, Verneuil-en-Halatte, France.,PERITOX UMR I-01 INERIS 01 UFR de médecine, Amiens, France
| | - Anne-Sophie Villégier
- Unité de Toxicologie Expérimentale, Institut National de l'Environnement Industriel et des Risques (INERIS), Parc Technologique ALATA, Verneuil-en-Halatte, France.,PERITOX UMR I-01 INERIS 01 UFR de médecine, Amiens, France
| |
Collapse
|
25
|
Tafakori S, Farrokhi A, Shalchyan V, Daliri MR. Investigating the impact of mobile range electromagnetic radiation on the medial prefrontal cortex of the rat during working memory. Behav Brain Res 2020; 391:112703. [PMID: 32461126 DOI: 10.1016/j.bbr.2020.112703] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2019] [Revised: 05/08/2020] [Accepted: 05/11/2020] [Indexed: 11/26/2022]
Abstract
Research has been focused on the effects of radiofrequency electromagnetic radiation (RF-EMR) emitted from a mobile phone on general health, especially the nervous system. The purpose of this study was to investigate the impact of RF-EMR on the brain mechanism of rats by recording local field potentials (LFPs) signals during working memory (WM) task. Subjects were exposed to 900 MHz from a dipole antenna daily for three hours. Exposure was applied, first on a short term base (1 week) and then on a long term base (4 weeks). Behavioral parameters were measured weekly while rats performed T-maze tasks in two types of normal and delayed. LFPs signals were simultaneously recorded by implanted microelectrode arrays on the medial prefrontal cortex (mPFC) of rats. Results showed a significant increase (*p < 0.05) in the task completion time of exposed rats which vanished shortly after the end of short term RF-EMR exposure. Before exposure, during correctly performed delayed tasks, an increase (peak) in power changes of theta band (4-12 Hz) was observed. But during correctly performed normal tasks, an increase appeared only by applying RF-EMR exposure. The similarity in power changes pattern of theta band in both types of tasks was observed after long term exposure. Classification accuracy of LFPs in truly done normal and delayed tasks was compared in pre and post-exposure states. Initial classification accuracy was 84.2 % which decreased significantly (*P < 0.05) after exposure. These observations indicated that RF-EMR may cause unusual brain functioning which is temporary at least for short term exposure.
Collapse
Affiliation(s)
- Shiva Tafakori
- Neuroscience and Neuroengineering Research Lab., Biomedical Engineering Department, School of Electrical Engineering, Iran University of Science and Technology (IUST), Narmak, 16846-13114, Tehran, Iran
| | - Ashkan Farrokhi
- Neuroscience and Neuroengineering Research Lab., Biomedical Engineering Department, School of Electrical Engineering, Iran University of Science and Technology (IUST), Narmak, 16846-13114, Tehran, Iran
| | - Vahid Shalchyan
- Neuroscience and Neuroengineering Research Lab., Biomedical Engineering Department, School of Electrical Engineering, Iran University of Science and Technology (IUST), Narmak, 16846-13114, Tehran, Iran.
| | - Mohammad Reza Daliri
- Neuroscience and Neuroengineering Research Lab., Biomedical Engineering Department, School of Electrical Engineering, Iran University of Science and Technology (IUST), Narmak, 16846-13114, Tehran, Iran.
| |
Collapse
|
26
|
Effects of a Single Head Exposure to GSM-1800 MHz Signals on the Transcriptome Profile in the Rat Cerebral Cortex: Enhanced Gene Responses Under Proinflammatory Conditions. Neurotox Res 2020; 38:105-123. [PMID: 32200527 PMCID: PMC7223958 DOI: 10.1007/s12640-020-00191-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Revised: 02/28/2020] [Accepted: 03/06/2020] [Indexed: 01/02/2023]
Abstract
Mobile communications are propagated by electromagnetic fields (EMFs), and since the 1990s, they operate with pulse-modulated signals such as the GSM-1800 MHz. The biological effects of GSM-EMF in humans affected by neuropathological processes remain seldom investigated. In this study, a 2-h head-only exposure to GSM-1800 MHz was applied to (i) rats undergoing an acute neuroinflammation triggered by a lipopolysaccharide (LPS) treatment, (ii) age-matched healthy rats, or (iii) transgenic hSOD1G93A rats that modeled a presymptomatic phase of human amyotrophic lateral sclerosis (ALS). Gene responses were assessed 24 h after the GSM head-only exposure in a motor area of the cerebral cortex (mCx) where the mean specific absorption rate (SAR) was estimated to be 3.22 W/kg. In LPS-treated rats, a genome-wide mRNA profiling was performed by RNA-seq analysis and revealed significant (adjusted p value < 0.05) but moderate (fold changes < 2) upregulations or downregulations affecting 2.7% of the expressed genes, including genes expressed predominantly in neuronal or in glial cell types and groups of genes involved in protein ubiquitination or dephosphorylation. Reverse transcription-quantitative PCR analyses confirmed gene modulations uncovered by RNA-seq data and showed that in a set of 15 PCR-assessed genes, significant gene responses to GSM-1800 MHz depended upon the acute neuroinflammatory state triggered in LPS-treated rats, because they were not observed in healthy or in hSOD1G93A rats. Together, our data specify the extent of cortical gene modulations triggered by GSM-EMF in the course of an acute neuroinflammation and indicate that GSM-induced gene responses can differ according to pathologies affecting the CNS.
Collapse
|
27
|
Stefi AL, Margaritis LH, Skouroliakou AS, Vassilacopoulou D. Mobile phone electromagnetic radiation affects Amyloid Precursor Protein and α-synuclein metabolism in SH-SY5Y cells. PATHOPHYSIOLOGY 2019; 26:203-212. [DOI: 10.1016/j.pathophys.2019.02.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Revised: 02/19/2019] [Accepted: 02/22/2019] [Indexed: 12/26/2022] Open
|
28
|
von Niederhäusern N, Ducray A, Zielinski J, Murbach M, Mevissen M. Effects of radiofrequency electromagnetic field exposure on neuronal differentiation and mitochondrial function in SH-SY5Y cells. Toxicol In Vitro 2019; 61:104609. [PMID: 31351122 DOI: 10.1016/j.tiv.2019.104609] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Revised: 07/23/2019] [Accepted: 07/23/2019] [Indexed: 01/08/2023]
Abstract
Exposure to radiofrequency electromagnetic fields (RF-EMF) has dramatically increased in the last decades with expanding use of mobile phones worldwide. The aim of this study was to evaluate effects of RF-EMF on neuronal differentiation and underlying signaling pathways involved in neuronal differentiation, neurodegeneration, and mitochondrial function. Differentiation of SH-SY5Y cells was performed using all-trans retinoic acid or staurosporine to obtain cholinergic and dopaminergic neurons. Exposure of SH-SY5Y cells at 935 MHz, 4 W/kg for 24 h did not alter the neuronal phenotypes quantitatively. Markers of the signaling pathways investigated, namely the mitogen-activated protein kinases (MAPK), extracellular signal-regulated kinases (Erk) 1 and 2 (p-Erk1/2) and protein kinase B (Akt), glycogen synthase kinase 3 β (GSK3β) and Wnt/β-catenin were not significantly affected by RF-EMF compared to sham. RF-EMF-impaired mitochondrial respiration in cells under glucose deprivation, but glutathione levels and mitochondrial fission and fusion markers were not altered. These findings indicate that RF-EMF might lead to an impairment of mitochondrial function that is only manifest at maximal respiration and additional stressors such as glucose deprivation. Further research is needed to investigate the effects of RF-EMF on mitochondrial function in detail because mitochondrial impairment is closely related to the pathogenesis of neurodegenerative diseases.
Collapse
Affiliation(s)
- Nicole von Niederhäusern
- Veterinary Pharmacology and Toxicology, Vetsuisse Faculty, University of Bern, Laenggassstrasse 124, 3012 Bern, Switzerland.
| | - Angélique Ducray
- Veterinary Pharmacology and Toxicology, Vetsuisse Faculty, University of Bern, Laenggassstrasse 124, 3012 Bern, Switzerland.
| | - Jana Zielinski
- Veterinary Pharmacology and Toxicology, Vetsuisse Faculty, University of Bern, Laenggassstrasse 124, 3012 Bern, Switzerland
| | - Manuel Murbach
- IT'IS Foundation, Zeughausstrasse 43, 8004 Zurich, Switzerland.
| | - Meike Mevissen
- Veterinary Pharmacology and Toxicology, Vetsuisse Faculty, University of Bern, Laenggassstrasse 124, 3012 Bern, Switzerland.
| |
Collapse
|
29
|
Tsoy A, Saliev T, Abzhanova E, Turgambayeva A, Kaiyrlykyzy A, Akishev M, Saparbayev S, Umbayev B, Askarova S. The Effects of Mobile Phone Radiofrequency Electromagnetic Fields on β-Amyloid-Induced Oxidative Stress in Human and Rat Primary Astrocytes. Neuroscience 2019; 408:46-57. [PMID: 30953670 DOI: 10.1016/j.neuroscience.2019.03.058] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Revised: 03/26/2019] [Accepted: 03/27/2019] [Indexed: 12/25/2022]
Abstract
Amyloid beta peptide (Aβ) is implicated in the development of pathological reactions associated with Alzheimer's disease (AD), such as oxidative stress, neuro-inflammation and death of brain cells. Current pharmacological approaches to treat AD are not able to control the deposition of Aβ and suppression of Aβ-induced cellular response. There is a growing body of evidence that exposure to radiofrequency electromagnetic field (RF-EMF) causes a decrease of beta-amyloid deposition in the brains and provides cognitive benefits to Alzheimer's Tg mice. Herein, we investigated the effects of mobile phone radiofrequency EMF of 918 MHz on reactive oxygen species (ROS) formation, mitochondrial membrane potential (MMP), activity of NADPH-oxidase, and phosphorylation of p38MAPK and ERK1/2 kinases in human and rat primary astrocytes in the presence of Aβ42 and H2O2. Our data demonstrate that EMF is able to reduce Aβ42- and H2O2-induced cellular ROS, abrogate Aβ₄₂-induced production of mitochondrial ROS and the co-localization between the cytosolic (p47-phox) and membrane (gp91-phox) subunits of NADPH oxidase, while increasing MMP, and inhibiting H2O2-induced phosphorylation of p38MAPK and ERK1/2 in primary astrocytes. Yet, EMF was not able to modulate alterations in the phosphorylation state of the MAPKs triggered by Aβ42. Our findings provide an insight into the mechanisms of cellular and molecular responses of astrocytes on RF-EMF exposure and indicate the therapeutic potential of RF-EMF for the treatment of Alzheimer's disease.
Collapse
Affiliation(s)
- Andrey Tsoy
- National Laboratory Astana, Nazarbayev University, 53 Kabanbay batyr Ave., Astana, 010000, Kazakhstan
| | - Timur Saliev
- National Laboratory Astana, Nazarbayev University, 53 Kabanbay batyr Ave., Astana, 010000, Kazakhstan; S.D. Asfendiyarov Kazakh National Medical University, Tole Bi Street 94, Almaty, 050000, Kazakhstan
| | - Elvira Abzhanova
- National Laboratory Astana, Nazarbayev University, 53 Kabanbay batyr Ave., Astana, 010000, Kazakhstan
| | - Anel Turgambayeva
- National Laboratory Astana, Nazarbayev University, 53 Kabanbay batyr Ave., Astana, 010000, Kazakhstan
| | - Aiym Kaiyrlykyzy
- National Laboratory Astana, Nazarbayev University, 53 Kabanbay batyr Ave., Astana, 010000, Kazakhstan
| | - Mars Akishev
- National Laboratory Astana, Nazarbayev University, 53 Kabanbay batyr Ave., Astana, 010000, Kazakhstan
| | - Samat Saparbayev
- National Scientific Medical Center, 42 Abylai Khan Ave, Astana, 010000, Kazakhstan, 010009
| | - Bauyrzhan Umbayev
- National Laboratory Astana, Nazarbayev University, 53 Kabanbay batyr Ave., Astana, 010000, Kazakhstan
| | - Sholpan Askarova
- National Laboratory Astana, Nazarbayev University, 53 Kabanbay batyr Ave., Astana, 010000, Kazakhstan.
| |
Collapse
|
30
|
Karimi SA, Salehi I, Shykhi T, Zare S, Komaki A. Effects of exposure to extremely low-frequency electromagnetic fields on spatial and passive avoidance learning and memory, anxiety-like behavior and oxidative stress in male rats. Behav Brain Res 2019; 359:630-638. [DOI: 10.1016/j.bbr.2018.10.002] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2018] [Revised: 09/14/2018] [Accepted: 10/01/2018] [Indexed: 12/11/2022]
|
31
|
Isoflurane preconditioning ameliorates electromagnetic pulse-induced neural damage by shifting microglia polarization toward anti-inflammatory phenotype via upregulation of SOCS1. Int Immunopharmacol 2019; 68:48-57. [PMID: 30611001 DOI: 10.1016/j.intimp.2018.12.064] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Revised: 12/04/2018] [Accepted: 12/28/2018] [Indexed: 01/20/2023]
Abstract
With the speedy technological advances during the past few decades, human exposure to the electromagnetic field (EMF) has become increasingly common. Exposure to EMF may induce neural injuries and dysfunction of various organs, likely involving neuroinflammation and activation of microglial cells. Isoflurane preconditioning (IP) is shown to provide neuroprotection in various neurological diseases by inhibiting excessive neuroinflammatory responses. Brain samples harvested from rats exposed to electromagnetic pulse (EMP) with or without IP were subjected to qPCR, Western blot assay, and immunohistochemistry to determine the expression of pro-inflammatory/anti-inflammatory microglia markers and a variety of pro- and anti-inflammatory mediators. Suppressor of cytokine signaling 1 (SOCS1) siRNA was used in cultured N9 microglia cells to examine the roles of SOCS1 in the effect of IP. In both in vivo and in vitro experiments, EMP-exposed microglia were predominantly pro-inflammatory microglia, accompanied by increased expression of pro-inflammatory cytokines and chemokines, and activation of TLR4 pathway, leading to neuronal death. IP reversed the changes induced by EMP and switched the activated microglia to an anti-inflammatory phenotype. SOCS1 siRNA abolished the beneficial effects of IP. IP ameliorates EMP-induced neural injuries by shifting microglia polarization from pro-inflammatory to anti-inflammatory phenotype via upregulation of SOCS1.
Collapse
|
32
|
Arendash G, Cao C, Abulaban H, Baranowski R, Wisniewski G, Becerra L, Andel R, Lin X, Zhang X, Wittwer D, Moulton J, Arrington J, Smith A. A Clinical Trial of Transcranial Electromagnetic Treatment in Alzheimer's Disease: Cognitive Enhancement and Associated Changes in Cerebrospinal Fluid, Blood, and Brain Imaging. J Alzheimers Dis 2019; 71:57-82. [PMID: 31403948 PMCID: PMC6839500 DOI: 10.3233/jad-190367] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/13/2019] [Indexed: 12/12/2022]
Abstract
BACKGROUND Small aggregates (oligomers) of the toxic proteins amyloid-β (Aβ) and phospho-tau (p-tau) are essential contributors to Alzheimer's disease (AD). In mouse models for AD or human AD brain extracts, Transcranial Electromagnetic Treatment (TEMT) disaggregates both Aβ and p-tau oligomers, and induces brain mitochondrial enhancement. These apparent "disease-modifying" actions of TEMT both prevent and reverse memory impairment in AD transgenic mice. OBJECTIVE To evaluate the safety and initial clinical efficacy of TEMT against AD, a comprehensive open-label clinical trial was performed. METHODS Eight mild/moderate AD patients were treated with TEMT in-home by their caregivers for 2 months utilizing a unique head device. TEMT was given for two 1-hour periods each day, with subjects primarily evaluated at baseline, end-of-treatment, and 2 weeks following treatment completion. RESULTS No deleterious behavioral effects, discomfort, or physiologic changes resulted from 2 months of TEMT, as well as no evidence of tumor or microhemorrhage induction. TEMT induced clinically important and statistically significant improvements in ADAS-cog, as well as in the Rey AVLT. TEMT also produced increases in cerebrospinal fluid (CSF) levels of soluble Aβ1-40 and Aβ1-42, cognition-related changes in CSF oligomeric Aβ, a decreased CSF p-tau/Aβ1-42 ratio, and reduced levels of oligomeric Aβ in plasma. Pre- versus post-treatment FDG-PET brain scans revealed stable cerebral glucose utilization, with several subjects exhibiting enhanced glucose utilization. Evaluation of diffusion tensor imaging (fractional anisotropy) scans in individual subjects provided support for TEMT-induced increases in functional connectivity within the cognitively-important cingulate cortex/cingulum. CONCLUSION TEMT administration to AD subjects appears to be safe, while providing cognitive enhancement, changes to CSF/blood AD markers, and evidence of stable/enhanced brain connectivity.
Collapse
Affiliation(s)
| | - Chuanhai Cao
- College of Pharmacy, University of South Florida, Tampa, FL, USA
| | - Haitham Abulaban
- University of South Florida Health/Byrd Alzheimer’s Institute, Tampa, FL, USA
| | | | | | | | - Ross Andel
- School of Aging Studies, University of South Florida, Tampa, FL, USA
- Department of Neurology, 2nd Faculty of Medicine, Charles University/Motol University Hospital, Prague, Czech Republic
| | - Xiaoyang Lin
- College of Pharmacy, University of South Florida, Tampa, FL, USA
| | - Xiaolin Zhang
- College of Pharmacy, University of South Florida, Tampa, FL, USA
| | | | | | | | - Amanda Smith
- University of South Florida Health/Byrd Alzheimer’s Institute, Tampa, FL, USA
| |
Collapse
|
33
|
Hidisoglu E, Kantar-Gok D, Ozen S, Yargicoglu P. Short-term 2.1 GHz radiofrequency radiation treatment induces significant changes on the auditory evoked potentials in adult rats. Int J Radiat Biol 2018; 94:858-871. [DOI: 10.1080/09553002.2018.1492166] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Affiliation(s)
- Enis Hidisoglu
- Faculty of Medicine, Department of Biophysics, Akdeniz University, Antalya, Turkey
| | - Deniz Kantar-Gok
- Faculty of Medicine, Department of Biophysics, Akdeniz University, Antalya, Turkey
| | - Sukru Ozen
- Engineering Faculty, Department of Electrical and Electronics Engineering, Akdeniz University, Antalya, Turkey
| | - Piraye Yargicoglu
- Faculty of Medicine, Department of Biophysics, Akdeniz University, Antalya, Turkey
| |
Collapse
|
34
|
Lian HY, Lin KW, Yang C, Cai P. Generation and propagation of yeast prion [URE3] are elevated under electromagnetic field. Cell Stress Chaperones 2018; 23:581-594. [PMID: 29214607 PMCID: PMC6045541 DOI: 10.1007/s12192-017-0867-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Revised: 11/24/2017] [Accepted: 11/27/2017] [Indexed: 12/13/2022] Open
Abstract
In this study, we studied the effect of 2.0 GHz radio frequency electromagnetic field (RF-EMF) and 50 Hz extremely low frequency electromagnetic field (ELF-EMF) exposure on prion generation and propagation using two budding yeast strains, NT64C and SB34, as model organisms. Under exposure to RF-EMF or ELF-EMF, the de novo generation and propagation of yeast prions [URE3] were elevated in both strains. The elevation increased over time, and the effects of ELF-EMF occurred in a dose-dependent manner. The transcription and expression levels of the molecular chaperones Hsp104, Hsp70-Ssa1/2, and Hsp40-Ydj1 were not statistically significantly changed after exposure. Furthermore, the levels of ROS, as well as the activities of superoxide dismutase (SOD) and catalase (CAT), were significantly elevated after short-term, but not long-term exposure. This work demonstrated for the first time that EMF exposure could elevate the de novo generation and propagation of yeast prions and supports the hypothesis that ROS may play a role in the effects of EMF on protein misfolding. The effects of EMF on protein folding and ROS levels may mediate the broad effects of EMF on cell function.
Collapse
Affiliation(s)
- Hui-Yong Lian
- Xiamen Key Laboratory of Physical Environment, Key Laboratory of Urban Environment and Health, Institute of Urban Environment, Chinese Academy of Sciences, 1799 Jimei Road, Xiamen, 361021, People's Republic of China.
- College of Life Sciences and Ecology, Hainan Tropical Ocean University, 1 Yucai Road, Sanya, 572022, People's Republic of China.
| | - Kang-Wei Lin
- Xiamen Key Laboratory of Physical Environment, Key Laboratory of Urban Environment and Health, Institute of Urban Environment, Chinese Academy of Sciences, 1799 Jimei Road, Xiamen, 361021, People's Republic of China
- University of the Chinese Academy of Sciences, 19 Yuquan Road, Beijing, 100049, People's Republic of China
| | - Chuanjun Yang
- Xiamen Key Laboratory of Physical Environment, Key Laboratory of Urban Environment and Health, Institute of Urban Environment, Chinese Academy of Sciences, 1799 Jimei Road, Xiamen, 361021, People's Republic of China
| | - Peng Cai
- Xiamen Key Laboratory of Physical Environment, Key Laboratory of Urban Environment and Health, Institute of Urban Environment, Chinese Academy of Sciences, 1799 Jimei Road, Xiamen, 361021, People's Republic of China.
| |
Collapse
|
35
|
Bevelacqua JJ, Mortazavi SMJ. Alzheimer 's Disease: Possible Mechanisms Behind Neurohormesis Induced by Exposure to Low Doses of Ionizing Radiation. J Biomed Phys Eng 2018; 8:153-156. [PMID: 29951441 PMCID: PMC6015644] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Accepted: 04/02/2018] [Indexed: 06/08/2023]
Abstract
In 2016, scientists reported that human exposure to low doses of ionizing radiation (CT scans of the brain) might relieve symptoms of both Alzheimer's disease (AD) and Parkinson disease (PD). The findings were unbelievable for those who were not familiar with neurohormesis. X-ray stimulation of the patient's adaptive protection systems against neurodegenerative diseases was the mechanism proposed by those authors. Now, some more recent studies performed in the field of neurobiological research confirm that low levels of stress can produce protective responses against the pathogenic processes. This paper outlines possible protective consequences of LDR in preventing the pathogenesis of AD through mechanisms such as restoring the myelin sheath and preventing neurodegeneration caused by oxidative stress. Focal demyelination is frequently reported in the proximity of beta-amyloid plaques within neocortex. Extracellular accumulation of amyloid is among well-characterized pathological changes in AD. It should be noted that LDR has been shown to contribute to the regeneration and functional recovery after transverse peripheral nerve injury (through inducing increased production of VEGF and GAP-43), which advances both the axonal regeneration and myelination. Another mechanism which is possibly involved is preventing neurodegeneration caused by oxidative stress. While high doses can induce reactive oxygen species (ROS) formation, oxidative stress and neuro-inflammation, substantial evidence now indicates that LDR can mitigate tissue damage through antioxidant defenses. Although adult neurogenesis has been reported to be beneficial for the regeneration of nervous system, some studies demonstrate that neurogenesis increases in AD brains. In spite of these reports, cellular therapy is introduced as a promising strategy for AD, and hence, LDR can affect the proliferation and differentiation of neural stem cells. Although such mechanisms are not fully known yet, it is hoped that this paper would foster further investigation into the mechanisms of this phenomenon, which accordingly improves human health.
Collapse
Affiliation(s)
- J J Bevelacqua
- Bevelacqua Resources, 343 Adair Drive, Richland, WA 99352, USA
| | - S M J Mortazavi
- Biophotonics Lab, Department of Electrical Engineering, University of Wisconsin Milwaukee, 3200 N Cramer St., Milwaukee, WI 53211, USA
- Ionizing and Non-ionizing Radiation Protection Research Center (INIRPRC), Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
36
|
Tekieh T, Sasanpour P, Rafii-Tabar H. Electrophysiological effects of low frequency electrical radiation on the neural compartment: a theoretical investigation. Biomed Phys Eng Express 2018. [DOI: 10.1088/2057-1976/aaa378] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
|
37
|
Curcio G. Exposure to Mobile Phone-Emitted Electromagnetic Fields and Human Attention: No Evidence of a Causal Relationship. Front Public Health 2018. [PMID: 29527523 PMCID: PMC5829032 DOI: 10.3389/fpubh.2018.00042] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
In the past 20 years of research regarding effects of mobile phone-derived electromagnetic fields (EMFs) on human cognition, attention has been one of the first and most extensively investigated functions. Different domains investigated covered selective, sustained, and divided attention. Here, the most relevant studies on this topic have been reviewed and discussed. A total of 43 studies are reported and summarized: of these, 31 indicated a total absence of statistically significant difference between real and sham signal, 9 showed a partial improvement of attentional performance (mainly increase in speed of performance and/or improvement of accuracy) as a function of real exposure, while the remaining 3 showed inconsistent results (i.e., increased speed in some tasks and slowing in others) or even a worsening in performance (reduced speed and/or deteriorated accuracy). These results are independent of the specific attentional domain investigated. This scenario allows to conclude that there is a substantial lack of evidence about a negative influence of non-ionizing radiations on attention functioning. Nonetheless, published literature is very heterogeneous under the point of view of methodology (type of signal, exposure time, blinding), dosimetry (accurate evaluation of specific absorption rate-SAR or emitted power), and statistical analyses, making arduous a conclusive generalization to everyday life. Some remarks and suggestions regarding future research are proposed.
Collapse
Affiliation(s)
- Giuseppe Curcio
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| |
Collapse
|
38
|
Park J, Kwon JH, Kim N, Song K. Effects of 1950 MHz radiofrequency electromagnetic fields on Aβ processing in human neuroblastoma and mouse hippocampal neuronal cells. JOURNAL OF RADIATION RESEARCH 2018; 59:18-26. [PMID: 29040655 PMCID: PMC5778507 DOI: 10.1093/jrr/rrx045] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/24/2017] [Revised: 05/31/2017] [Indexed: 06/07/2023]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disease leading to progressive loss of memory and other cognitive functions. One of the well-known pathological markers of AD is the accumulation of amyloid-beta protein (Aβ), and its plaques, in the brain. Recent studies using Tg-5XFAD mice as a model of AD have reported that exposure to radiofrequency electromagnetic fields (RF-EMF) from cellular phones reduced Aβ plaques in the brain and showed beneficial effects on AD. In this study, we examined whether exposure to 1950 MHz RF-EMF affects Aβ processing in neural cells. We exposed HT22 mouse hippocampal neuronal cells and SH-SY5Y human neuroblastoma cells to RF-EMF (SAR 6 W/kg) for 2 h per day for 3 days, and analyzed the mRNA and protein expression of the key genes related to Aβ processing. When exposed to RF-EMF, mRNA levels of APP, BACE1, ADAM10 and PSEN1 were decreased in HT22, but the mRNA level of APP was not changed in SH-SY5Y cells. The protein expression of APP and BACE1, as well as the secreted Aβ peptide, was not significantly different between RF-EMF-exposed 7w-PSML, HT22 and SH-SY5Y cells and the unexposed controls. These observations suggest that RF-EMF exposure may not have a significant physiological effect on Aβ processing of neural cells in the short term. However, considering that we only exposed HT22 and SH-SY5Y cells to RF-EMF for 2 h per day for 3 days, we cannot exclude the possibility that 1950 MHz RF-EMF induces physiological change in Aβ processing with long-term and continuous exposure.
Collapse
Affiliation(s)
- Jeongyeon Park
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul 03722, Republic of Korea
| | - Jong Hwa Kwon
- Department of EMF Research Team, Radio and Broadcasting Technology Laboratory, Electronics and Telecommunications Research Institute (ETRI), Daejon, 305-700, Republic of Korea
| | - Nam Kim
- School of Electrical and Computer Engineering, Chungbuk National University, Cheongju, Chungbuk, 362-763, Republic of Korea
| | - Kiwon Song
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul 03722, Republic of Korea
| |
Collapse
|
39
|
Son Y, Kim JS, Jeong YJ, Jeong YK, Kwon JH, Choi HD, Pack JK, Kim N, Lee YS, Lee HJ. Long-term RF exposure on behavior and cerebral glucose metabolism in 5xFAD mice. Neurosci Lett 2017; 666:64-69. [PMID: 29273398 DOI: 10.1016/j.neulet.2017.12.042] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Revised: 12/14/2017] [Accepted: 12/18/2017] [Indexed: 11/25/2022]
Abstract
Given the increased public concern about the deleterious biological consequences of radiofrequency electromagnetic fields (RF-EMFs), the involvement of RF-EMFs in neurodegenerative diseases, especially Alzheimer's disease (AD), has received increased consideration. To investigate the effect of long-term RF-EMF exposure on AD progression, we exposed 5xFAD mice to 1950 MHz RF-EMF at a specific absorption rate of 5.0 W/kg for 2 h/day and 5 days/week for 8 months. Behavioral changes were assessed by an open field test and an object recognition memory task after RF exposure was terminated. In addition, cerebral glucose metabolism was analyzed in the brains of the 5xFAD mice using 18F-deoxyglucose positron emission tomography. The hyperactivity-like and anxiolytic behaviors of the 5xFAD mice in open field tests were rescued by RF exposure. Furthermore, long-term RF-EMF exposure improved the cognitive deficits of 5xFAD mice that were observed in the object recognition memory test. Consistent with the behavioral changes, glucose metabolism in the hippocampus and amygdala regions of the brains of 5xFAD mice following RF exposure was significantly increased compared to glucose metabolism in the brains of sham-exposed mice. These data suggest that long-term exposure to RF-EMF might exert beneficial effects on AD in 5xFAD mice.
Collapse
Affiliation(s)
- Yeonghoon Son
- Division of Basic Radiation Bioscience, Korea Institute of Radiological & Medical Sciences, Seoul, Republic of Korea; National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju, Republic of Korea
| | - Jin Su Kim
- Division of RI Research, Korea Institute of Radiological & Medical Sciences, Seoul, Republic of Korea
| | - Ye Ji Jeong
- Division of Basic Radiation Bioscience, Korea Institute of Radiological & Medical Sciences, Seoul, Republic of Korea
| | - Youn Kyoung Jeong
- Radiation Non-Clinic Center, Korea Institute of Radiological & Medical Sciences, Seoul, Republic of Korea
| | - Jong Hwa Kwon
- Department of EMF Research Team, Radio and Broadcasting Technology Laboratory, ETRI, Daejon, Republic of Korea
| | - Hyung-Do Choi
- Department of EMF Research Team, Radio and Broadcasting Technology Laboratory, ETRI, Daejon, Republic of Korea
| | - Jeong-Ki Pack
- Department of Radio Sciences and Engineering, College of Engineering, Chungnam National University, Daejon, Republic of Korea
| | - Nam Kim
- School of Electrical and Computer Engineering, Chungbuk National University, Cheongju, Chungbuk, Republic of Korea
| | - Yun-Sil Lee
- Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul, Republic of Korea.
| | - Hae-June Lee
- Division of Basic Radiation Bioscience, Korea Institute of Radiological & Medical Sciences, Seoul, Republic of Korea.
| |
Collapse
|
40
|
Sakhaie MH, Soleimani M, Pourheydar B, Majd Z, Atefimanesh P, Asl SS, Mehdizadeh M. Effects of Extremely Low-Frequency Electromagnetic Fields on Neurogenesis and Cognitive Behavior in an Experimental Model of Hippocampal Injury. Behav Neurol 2017; 2017:9194261. [PMID: 29259353 PMCID: PMC5702423 DOI: 10.1155/2017/9194261] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Revised: 07/23/2017] [Accepted: 08/08/2017] [Indexed: 01/19/2023] Open
Abstract
Exposure to extremely low-frequency electromagnetic fields may induce constant modulation in neuronal plasticity. In recent years, tremendous efforts have been made to design a suitable strategy for enhancing adult neurogenesis, which seems to be deterred due to brain senescence and several neurodegenerative diseases. In this study, we evaluated the effects of ELF-EMF on neurogenesis and memory, following treatment with trimethyltin chloride (TMT) as a neurotoxicant. The mice in all groups (n = 56) were injected with BrdU during the experiment for seven consecutive days to label newborn cells. Spatial memory was assessed by the Morris water maze (MWM) test. By the end of the experiment, neurogenesis and neuronal differentiation were assessed in the hippocampus, using immunohistochemistry and Western blot analysis. Based on the findings, exposure to ELF-EMF enhanced spatial learning and memory in the MWM test. ELF-EMF exposure significantly enhanced the number of BrdU+ and NeuN+ cells in the dentate gyrus of adult mice (P < 0.001 and P < 0.05, resp.). Western blot analysis revealed significant upregulation of NeuroD2 in ELF-EMF-exposed mice compared to the TMT-treated group (P < 0.05). These findings suggest that ELF-EMF might have clinical implications for the improvement of neurodegenerative processes and could help develop a novel therapeutic approach in regenerative medicine.
Collapse
Affiliation(s)
- Mohammad Hassan Sakhaie
- Cellular and Molecular Research Center and Department of Anatomy, Iran University of Medical Sciences, Tehran, Iran
- Department of Anatomy, Arak University of Medical Sciences, Arak, Iran
| | - Mansoureh Soleimani
- Cellular and Molecular Research Center and Department of Anatomy, Iran University of Medical Sciences, Tehran, Iran
| | - Bagher Pourheydar
- Urmia University of Medical Sciences, Faculty of Medicine, Neurophysiology Research Center, Department of Anatomy, Urmia, Iran
| | - Zahra Majd
- Oncopathology Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Pezhman Atefimanesh
- Cellular and Molecular Research Center, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Sara Soleimani Asl
- Endometrium and Endometriosis Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
- Anatomy Department, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Mehdi Mehdizadeh
- Cellular and Molecular Research Center, Faculty of Advanced Technologies in Medicine, Department of Anatomy, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
41
|
Deniz OG, Kaplan S, Selçuk MB, Terzi M, Altun G, Yurt KK, Aslan K, Davis D. Effects of short and long term electromagnetic fields exposure on the human hippocampus. J Microsc Ultrastruct 2017; 5:191-197. [PMID: 30023254 PMCID: PMC6025790 DOI: 10.1016/j.jmau.2017.07.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Revised: 06/05/2017] [Accepted: 07/02/2017] [Indexed: 12/23/2022] Open
Abstract
The increasing use of mobile phones may have a number of physiological and psychological effects on human health. Many animal and human studies have reported various effects on the central nervous system and cognitive performance from of exposure to electromagnetic fields (EMF) emitted by mobile phones. The aim of the present study was to evaluate the effects of mobile phones on the morphology of the human brain and on cognitive performance using stereological and spectroscopic methods and neurocognitive tests. Sixty healthy female medical school students aged 18-25 years were divided into a low exposure group (30 subjects, <30 min daily use by the head) and high exposure group (30 subjects, >90 min daily use by the head). Magnetic resonance images (MRI) of the brain analysed on OsiriX 3.2.1 workstation. Neuropsychological tests were performed for each subject. In addition, three dominant specific metabolites were analysed, choline at 3.21 ppm, creatine at 3.04 ppm and N-acetyl aspartate at 2.02 ppm. Analysis of the spectroscopic results revealed no significant difference in specific metabolites between the groups (p > 0.05). There was also no significant difference in terms of hippocampal volume between the groups (p > 0.05). In contrast, the results of the stroop and digit span (backward) neurocognitive tests of high exposure group for evaluating attention were significantly poorer from low exposure group (p < 0.05). Based on these results, we conclude that a lack of attention and concentration may occur in subjects who talk on mobile phones for longer times, compared to those who use phones relatively less.
Collapse
Affiliation(s)
- Omur Gulsum Deniz
- Department of Histology and Embryology, Medical School of Ondokuz Mayıs University, Samsun, Turkey
| | - Suleyman Kaplan
- Department of Histology and Embryology, Medical School of Ondokuz Mayıs University, Samsun, Turkey
| | - Mustafa Bekir Selçuk
- Department of Radiology, Medical School of Ondokuz Mayıs University, Samsun, Turkey
| | - Murat Terzi
- Department of Neurology, Medical School of Ondokuz Mayıs University, Samsun, Turkey
| | - Gamze Altun
- Department of Histology and Embryology, Medical School of Ondokuz Mayıs University, Samsun, Turkey
| | - Kıymet Kübra Yurt
- Department of Histology and Embryology, Medical School of Ondokuz Mayıs University, Samsun, Turkey
| | - Kerim Aslan
- Department of Radiology, Medical School of Ondokuz Mayıs University, Samsun, Turkey
| | - Devra Davis
- Department of Medicine and Public Health, The Hebrew University, Jerusalem, Israel
- Environmental Health Trust, Teton Village, WY, USA
| |
Collapse
|
42
|
Lameth J, Gervais A, Colin C, Lévêque P, Jay TM, Edeline JM, Mallat M. Acute Neuroinflammation Promotes Cell Responses to 1800 MHz GSM Electromagnetic Fields in the Rat Cerebral Cortex. Neurotox Res 2017; 32:444-459. [PMID: 28578480 DOI: 10.1007/s12640-017-9756-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Revised: 05/16/2017] [Accepted: 05/19/2017] [Indexed: 01/08/2023]
Abstract
Mobile phone communications are conveyed by radiofrequency (RF) electromagnetic fields, including pulse-modulated global system for mobile communications (GSM)-1800 MHz, whose effects on the CNS affected by pathological states remain to be specified. Here, we investigated whether a 2-h head-only exposure to GSM-1800 MHz could impact on a neuroinflammatory reaction triggered by lipopolysaccharide (LPS) in 2-week-old or adult rats. We focused on the cerebral cortex in which the specific absorption rate (SAR) of RF averaged 2.9 W/kg. In developing rats, 24 h after GSM exposure, the levels of cortical interleukin-1ß (IL1ß) or NOX2 NADPH oxidase transcripts were reduced by 50 to 60%, in comparison with sham-exposed animals (SAR = 0), as assessed by RT-qPCR. Adult rats exposed to GSM also showed a 50% reduction in the level of IL1ß mRNA, but they differed from developing rats by the lack of NOX2 gene suppression and by displaying a significant growth response of microglial cell processes imaged in anti-Iba1-stained cortical sections. As neuroinflammation is often associated with changes in excitatory neurotransmission, we evaluated changes in expression and phosphorylation of α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptors in the adult cerebral cortex by Western blot analyses. We found that GSM exposure decreased phosphorylation at two residues on the GluA1 AMPAR subunit (serine 831 and 845). The GSM-induced changes in gene expressions, microglia, and GluA1 phosphorylation did not persist 72 h after RF exposure and were not observed in the absence of LPS pretreatment. Together, our data provide evidence that GSM-1800 MHz can modulate CNS cell responses triggered by an acute neuroinflammatory state.
Collapse
Affiliation(s)
- Julie Lameth
- Sorbonne Universités, UPMC Univ Paris 06, INSERM U.1127, CNRS, Institut du Cerveau et de la Moelle épinière (ICM), Hôpital Pitié-Salpêtrière, Bat. ICM, 47 boulevard de l'Hôpital, F-75013, Paris, France
| | - Annie Gervais
- Sorbonne Universités, UPMC Univ Paris 06, INSERM U.1127, CNRS, Institut du Cerveau et de la Moelle épinière (ICM), Hôpital Pitié-Salpêtrière, Bat. ICM, 47 boulevard de l'Hôpital, F-75013, Paris, France
| | - Catherine Colin
- Sorbonne Universités, UPMC Univ Paris 06, INSERM U.1127, CNRS, Institut du Cerveau et de la Moelle épinière (ICM), Hôpital Pitié-Salpêtrière, Bat. ICM, 47 boulevard de l'Hôpital, F-75013, Paris, France
| | - Philippe Lévêque
- Université de Limoges, CNRS, XLIM, UMR 7252, 123 avenue Albert Thomas, F-87000, Limoges, France
| | - Thérèse M Jay
- Physiopathologie des Maladies Psychiatriques, Centre de Psychiatrie et Neurosciences, UMR_S894 INSERM, Université Paris Descartes, 102-108 rue de la Santé, 75014, Paris, France
| | - Jean-Marc Edeline
- Paris Saclay Institute of Neuroscience, Neuro-PSI, UMR 9197 CNRS, Université Paris-Sud, 91405, Orsay cedex, France
| | - Michel Mallat
- Sorbonne Universités, UPMC Univ Paris 06, INSERM U.1127, CNRS, Institut du Cerveau et de la Moelle épinière (ICM), Hôpital Pitié-Salpêtrière, Bat. ICM, 47 boulevard de l'Hôpital, F-75013, Paris, France.
| |
Collapse
|
43
|
Van Eeghem V, El Arfani A, Anthoula A, Walrave L, Pourkazemi A, Bentea E, Demuyser T, Smolders I, Stiens J. Selective changes in locomotor activity in mice due to low-intensity microwaves amplitude modulated in the EEG spectral domain. Neuroscience 2017; 359:40-48. [PMID: 28687311 DOI: 10.1016/j.neuroscience.2017.06.056] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Revised: 06/14/2017] [Accepted: 06/27/2017] [Indexed: 11/19/2022]
Abstract
Despite the numerous benefits of microwave applications in our daily life, microwaves were associated with diverse neurological complaints such as headaches and impaired sleep patterns, and changes in the electroencephalogram (EEG). To which extent microwaves influence the brain function remains unclear. This exploratory study assessed the behavior and neurochemistry in mice immediately or 4weeks after a 6-day exposure to low-intensity 10-GHz microwaves with an amplitude modulation (AM) of 2 or 8Hz. These modulation frequencies of 2 and 8Hz are situated within the delta and theta-alpha frequency bands in the EEG spectrum and are associated with sleep and active behavior, respectively. During these experiments, the specific absorbance rate was 0.3W/kg increasing the brain temperature with 0.23°C. For the first time, exposing mice to 8-Hz AM significantly reduced locomotor activity in an open field immediately after exposure which normalized after 4weeks. This in contrast to 2-Hz AM which didn't induce significant changes in locomotor activity immediately and 4weeks after exposure. Despite this difference in motor behavior, no significant changes in striatal dopamine (DA) and 3,4-dihydroxyphenylacetic acid (DOPAC) levels and DOPAC/DA turnover nor in cortical glutamate (GLU) concentrations were detected. In all cases, no effects on motor coordination on a rotarod, spatial working memory, anxiety nor depressive-like behavior were observed. The outcome of this study indicates that exposing mice to low-intensity 8-Hz AM microwaves can alter the locomotor activity in contrast to 2-Hz AM which did not affect the tested behaviors.
Collapse
Affiliation(s)
- Vincent Van Eeghem
- Department of Electronics and Informatics (ETRO), Laboratory for micro- and photon electronics (LAMI), Vrije Universiteit Brussel, Pleinlaan 2, 1050 Brussels, Belgium.
| | - Anissa El Arfani
- Laboratory of Pharmaceutical Chemistry, Drug Analysis and Drug Information (FASC), Research Group Experimental Pharmacology, Center for Neurosciences (C4N), Vrije Universiteit Brussel, Laarbeeklaan 103, 1090 Brussels, Belgium
| | - Arta Anthoula
- Department of Electronics and Informatics (ETRO), Laboratory for micro- and photon electronics (LAMI), Vrije Universiteit Brussel, Pleinlaan 2, 1050 Brussels, Belgium
| | - Laura Walrave
- Laboratory of Pharmaceutical Chemistry, Drug Analysis and Drug Information (FASC), Research Group Experimental Pharmacology, Center for Neurosciences (C4N), Vrije Universiteit Brussel, Laarbeeklaan 103, 1090 Brussels, Belgium
| | - Ali Pourkazemi
- Department of Electronics and Informatics (ETRO), Laboratory for micro- and photon electronics (LAMI), Vrije Universiteit Brussel, Pleinlaan 2, 1050 Brussels, Belgium
| | - Eduard Bentea
- Laboratory of Pharmaceutical Chemistry, Drug Analysis and Drug Information (FASC), Research Group Experimental Pharmacology, Center for Neurosciences (C4N), Vrije Universiteit Brussel, Laarbeeklaan 103, 1090 Brussels, Belgium
| | - Thomas Demuyser
- Laboratory of Pharmaceutical Chemistry, Drug Analysis and Drug Information (FASC), Research Group Experimental Pharmacology, Center for Neurosciences (C4N), Vrije Universiteit Brussel, Laarbeeklaan 103, 1090 Brussels, Belgium
| | - Ilse Smolders
- Laboratory of Pharmaceutical Chemistry, Drug Analysis and Drug Information (FASC), Research Group Experimental Pharmacology, Center for Neurosciences (C4N), Vrije Universiteit Brussel, Laarbeeklaan 103, 1090 Brussels, Belgium
| | - Johan Stiens
- Department of Electronics and Informatics (ETRO), Laboratory for micro- and photon electronics (LAMI), Vrije Universiteit Brussel, Pleinlaan 2, 1050 Brussels, Belgium.
| |
Collapse
|
44
|
Barthélémy A, Mouchard A, Bouji M, Blazy K, Puigsegur R, Villégier AS. Glial markers and emotional memory in rats following acute cerebral radiofrequency exposures. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2016; 23:25343-25355. [PMID: 27696165 DOI: 10.1007/s11356-016-7758-y] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Accepted: 09/21/2016] [Indexed: 06/06/2023]
Abstract
The widespread mobile phone use raises concerns on the possible cerebral effects of radiofrequency electromagnetic fields (RF EMF). Reactive astrogliosis was reported in neuroanatomical structures of adaptive behaviors after a single RF EMF exposure at high specific absorption rate (SAR, 6 W/kg). Here, we aimed to assess if neuronal injury and functional impairments were related to high SAR-induced astrogliosis. In addition, the level of beta amyloid 1-40 (Aβ 1-40) peptide was explored as a possible toxicity marker. Sprague Dawley male rats were exposed for 15 min at 0, 1.5, or 6 W/kg or for 45 min at 6 W/kg. Memory, emotionality, and locomotion were tested in the fear conditioning, the elevated plus maze, and the open field. Glial fibrillary acidic protein (GFAP, total and cytosolic fractions), myelin basic protein (MBP), and Aβ1-40 were quantified in six brain areas using enzyme-linked immunosorbent assay. According to our data, total GFAP was increased in the striatum (+114 %) at 1.5 W/kg. Long-term memory was reduced, and cytosolic GFAP was increased in the hippocampus (+119 %) and in the olfactory bulb (+46 %) at 6 W/kg (15 min). No MBP or Aβ1-40 expression modification was shown. Our data corroborates previous studies indicating RF EMF-induced astrogliosis. This study suggests that RF EMF-induced astrogliosis had functional consequences on memory but did not demonstrate that it was secondary to neuronal damage.
Collapse
Affiliation(s)
- Amélie Barthélémy
- Institut national de l'environnement industriel et des risques (INERIS), Unité de Toxicologie Expérimentale, Parc Technologique ALATA, BP no. 2, 60550, Verneuil-en-Halatte, France
- Institut des Neurosciences Cellulaires et Intégratives, CNRS UPR 3212, 5 rue Blaise Pascal, 67084, Strasbourg, France
| | - Amandine Mouchard
- Institut national de l'environnement industriel et des risques (INERIS), Unité de Toxicologie Expérimentale, Parc Technologique ALATA, BP no. 2, 60550, Verneuil-en-Halatte, France
- Institut des Maladies Neurodégénératives CNRS UMR5293 Université de Bordeaux, 146 rue Léo Saignat, 33076, Bordeaux, France
| | - Marc Bouji
- Institut national de l'environnement industriel et des risques (INERIS), Unité de Toxicologie Expérimentale, Parc Technologique ALATA, BP no. 2, 60550, Verneuil-en-Halatte, France
- Campus des sciences et technologies, Université Saint-Joseph, Dekwaneh, Mar Roukos, Lebanon
| | - Kelly Blazy
- Institut national de l'environnement industriel et des risques (INERIS), Unité de Toxicologie Expérimentale, Parc Technologique ALATA, BP no. 2, 60550, Verneuil-en-Halatte, France
- Unité mixte PERITOX EA 4285-UM INERIS 01 Laboratoire Périnatalité et risques toxicologiques CHU Amiens-Picardie Hôpital, Sud Avenue Laënnec, 80 480, Salouël, France
| | - Renaud Puigsegur
- Institut national de l'environnement industriel et des risques (INERIS), Unité de Toxicologie Expérimentale, Parc Technologique ALATA, BP no. 2, 60550, Verneuil-en-Halatte, France
- Sous-direction de la police technique et scientifique, 31 Avenue Franklin Roosevelt, 69130, Ecully, France
| | - Anne-Sophie Villégier
- Institut national de l'environnement industriel et des risques (INERIS), Unité de Toxicologie Expérimentale, Parc Technologique ALATA, BP no. 2, 60550, Verneuil-en-Halatte, France.
- Unité mixte PERITOX EA 4285-UM INERIS 01 Laboratoire Périnatalité et risques toxicologiques CHU Amiens-Picardie Hôpital, Sud Avenue Laënnec, 80 480, Salouël, France.
| |
Collapse
|
45
|
Son Y, Jeong YJ, Kwon JH, Choi H, Pack J, Kim N, Lee Y, Lee H. 1950 MHz radiofrequency electromagnetic fields do not aggravate memory deficits in 5xFAD mice. Bioelectromagnetics 2016; 37:391-9. [PMID: 27434853 PMCID: PMC5108492 DOI: 10.1002/bem.21992] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Accepted: 07/02/2016] [Indexed: 12/14/2022]
Abstract
The increased use of mobile phones has generated public concern about the impact of radiofrequency electromagnetic fields (RF-EMF) on health. In the present study, we investigated whether RF-EMFs induce molecular changes in amyloid precursor protein (APP) processing and amyloid beta (Aβ)-related memory impairment in the 5xFAD mouse, which is a widely used amyloid animal model. The 5xFAD mice at the age of 1.5 months were assigned to two groups (RF-EMF- and sham-exposed groups, eight mice per group). The RF-EMF group was placed in a reverberation chamber and exposed to 1950 MHz electromagnetic fields for 3 months (SAR 5 W/kg, 2 h/day, 5 days/week). The Y-maze, Morris water maze, and novel object recognition memory test were used to evaluate spatial and non-spatial memory following 3-month RF-EMF exposure. Furthermore, Aβ deposition and APP and carboxyl-terminal fragment β (CTFβ) levels were evaluated in the hippocampus and cortex of 5xFAD mice, and plasma levels of Aβ peptides were also investigated. In behavioral tests, mice that were exposed to RF-EMF for 3 months did not exhibit differences in spatial and non-spatial memory compared to the sham-exposed group, and no apparent change was evident in locomotor activity. Consistent with behavioral data, RF-EMF did not alter APP and CTFβ levels or Aβ deposition in the brains of the 5xFAD mice. These findings indicate that 3-month RF-EMF exposure did not affect Aβ-related memory impairment or Aβ accumulation in the 5xFAD Alzheimer's disease model. Bioelectromagnetics. 37:391-399, 2016. © 2016 The Authors Bioelectromagnetics published by Wiley Periodicals, Inc. on behalf of Bioelectromagnetics Society.
Collapse
Affiliation(s)
- Yeonghoon Son
- Division of Radiation EffectsKorea Institute of Radiological and Medical SciencesSeoulKorea
| | - Ye Ji Jeong
- Division of Radiation EffectsKorea Institute of Radiological and Medical SciencesSeoulKorea
| | - Jong Hwa Kwon
- Radio and Broadcasting Technology LaboratoryDepartment of EMF Research TeamDaejonKorea
| | - Hyung‐Do Choi
- Radio and Broadcasting Technology LaboratoryDepartment of EMF Research TeamDaejonKorea
| | - Jeong‐Ki Pack
- Department of Radio Sciences and EngineeringCollege of EngineeringChungnam National UniversityDaejonKorea
| | - Nam Kim
- School of Electrical and Computer EngineeringChungbuk National UniversityCheongjuChungbukKorea
| | - Yun‐Sil Lee
- Graduate School of Pharmaceutical SciencesEwha Womans UniversitySeoulKorea
| | - Hae‐June Lee
- Division of Radiation EffectsKorea Institute of Radiological and Medical SciencesSeoulKorea
| |
Collapse
|
46
|
Jiang DP, Li JH, Zhang J, Xu SL, Kuang F, Lang HY, Wang YF, An GZ, Li J, Guo GZ. Long-term electromagnetic pulse exposure induces Abeta deposition and cognitive dysfunction through oxidative stress and overexpression of APP and BACE1. Brain Res 2016; 1642:10-19. [DOI: 10.1016/j.brainres.2016.02.053] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2015] [Revised: 02/05/2016] [Accepted: 02/14/2016] [Indexed: 11/30/2022]
|
47
|
Arendash GW. Review of the Evidence that Transcranial Electromagnetic Treatment will be a Safe and Effective Therapeutic Against Alzheimer's Disease. J Alzheimers Dis 2016; 53:753-71. [PMID: 27258417 PMCID: PMC4981900 DOI: 10.3233/jad-160165] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/18/2016] [Indexed: 12/21/2022]
Abstract
We have demonstrated in multiple studies that daily, long-term electromagnetic field (EMF) treatment in the ultra-high frequency range not only protects Alzheimer's disease (AD) transgenic mice from cognitive impairment, but also reverses such impairment in aged AD mice. Moreover, these beneficial cognitive effects appear to be through direct actions on the AD process. Based on a large array of pre-clinical data, we have initiated a pilot clinical trial to determine the safety and efficacy of EMF treatment to mild-moderate AD subjects. Since it is important to establish the safety of this new neuromodulatory approach, the main purpose of this review is to provide a comprehensive assessment of evidence supporting the safety of EMFs, particularly through transcranial electromagnetic treatment (TEMT). In addition to our own pre-clinical studies, a rich variety of both animal and cell culture studies performed by others have underscored the anticipated safety of TEMT in clinical AD trials. Moreover, numerous clinical studies have determined that short- or long-term human exposure to EMFs similar to those to be provided clinically by TEMT do not have deleterious effects on general health, cognitive function, or a variety of physiologic measures-to the contrary, beneficial effects on brain function/activity have been reported. Importantly, such EMF exposure has not been shown to increase the risk of any type of cancer in human epidemiologic studies, as well as animal and cell culture studies. In view of all the above, clinical trials of safety/efficacy with TEMT to AD subjects are clearly warranted and now in progress.
Collapse
|
48
|
The role of electromagnetic fields in neurological disorders. J Chem Neuroanat 2016; 77:78-79. [PMID: 27126876 DOI: 10.1016/j.jchemneu.2016.04.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Accepted: 04/22/2016] [Indexed: 11/23/2022]
|
49
|
Schiavone S, Trabace L. Pharmacological targeting of redox regulation systems as new therapeutic approach for psychiatric disorders: A literature overview. Pharmacol Res 2016; 107:195-204. [PMID: 26995306 DOI: 10.1016/j.phrs.2016.03.019] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2016] [Revised: 03/14/2016] [Accepted: 03/14/2016] [Indexed: 12/20/2022]
Abstract
Redox dysregulation occurs following a disequilibrium between reactive oxygen species (ROS) producing and degrading systems, i.e. mitochondria, nicotinamide adenine dinucleotide phosphate (NADPH) oxidases and nitric oxide synthase (NOS) on one hand and the principal antioxidant system, the glutathione, on the other hand. Increasing recent evidence points towards a pathogenetic role of an altered redox state in the development of several mental disorders, such as anxiety, bipolar disorders, depression, psychosis, autism and post-traumaticstress disorders (PTSD). In this regard, pharmacological targeting of the redox state regulating systems in the brain has been proposed as an innovative and promising therapeutic approach for the treatment of these mental diseases. This review will summarize current knowledge obtained from both pre-clinical and clinical studies in order to descant "lights and shadows" of targeting pharmacologically both the producing and degrading reactive oxygen species (ROS) systems in psychiatric disorders.
Collapse
Affiliation(s)
- Stefania Schiavone
- Department of Clinical and Experimental Medicine, University of Foggia, Via Napoli, 20 71122 Foggia, Italy.
| | - Luigia Trabace
- Department of Clinical and Experimental Medicine, University of Foggia, Via Napoli, 20 71122 Foggia, Italy.
| |
Collapse
|
50
|
Radiofréquences et santé. ARCH MAL PROF ENVIRO 2015. [DOI: 10.1016/j.admp.2015.03.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|