1
|
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
|
2
|
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
|
3
|
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
|
4
|
Zhi W, Li Y, Wang L, Hu X. Advancing Neuroscience and Therapy: Insights into Genetic and Non-Genetic Neuromodulation Approaches. Cells 2025; 14:122. [PMID: 39851550 PMCID: PMC11763439 DOI: 10.3390/cells14020122] [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: 12/12/2024] [Revised: 12/31/2024] [Accepted: 01/07/2025] [Indexed: 01/26/2025] Open
Abstract
Neuromodulation stands as a cutting-edge approach in the fields of neuroscience and therapeutic intervention typically involving the regulation of neural activity through physical and chemical stimuli. The purpose of this review is to provide an overview and evaluation of different neuromodulation techniques, anticipating a clearer understanding of the future developmental trajectories and the challenges faced within the domain of neuromodulation that can be achieved. This review categorizes neuromodulation techniques into genetic neuromodulation methods (including optogenetics, chemogenetics, sonogenetics, and magnetogenetics) and non-genetic neuromodulation methods (including deep brain stimulation, transcranial magnetic stimulation, transcranial direct current stimulation, transcranial ultrasound stimulation, photobiomodulation therapy, infrared neuromodulation, electromagnetic stimulation, sensory stimulation therapy, and multi-physical-factor stimulation techniques). By systematically evaluating the principles, mechanisms, advantages, limitations, and efficacy in modulating neuronal activity and the potential applications in interventions of neurological disorders of these neuromodulation techniques, a comprehensive picture is gradually emerging regarding the advantages and challenges of neuromodulation techniques, their developmental trajectory, and their potential clinical applications. This review highlights significant advancements in applying these techniques to treat neurological and psychiatric disorders. Genetic methods, such as sonogenetics and magnetogenetics, have demonstrated high specificity and temporal precision in targeting neuronal populations, while non-genetic methods, such as transcranial magnetic stimulation and photobiomodulation therapy, offer noninvasive and versatile clinical intervention options. The transformative potential of these neuromodulation techniques in neuroscience research and clinical practice is underscored, emphasizing the need for integration and innovation in technologies, the optimization of delivery methods, the improvement of mediums, and the evaluation of toxicity to fully harness their therapeutic potential.
Collapse
Affiliation(s)
- Weijia Zhi
- Beijing Institute of Radiation Medicine, Beijing 100850, China;
| | - Ying Li
- School of Life Science and Technology, Xi’an Jiaotong University, Xi’an 710049, China;
| | - Lifeng Wang
- Beijing Institute of Radiation Medicine, Beijing 100850, China;
| | - Xiangjun Hu
- Beijing Institute of Radiation Medicine, Beijing 100850, China;
| |
Collapse
|
5
|
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
|
6
|
Perez FP, Morisaki J, Kanakri H, Rizkalla M. Electromagnetic Field Stimulation Therapy for Alzheimer's Disease. NEUROLOGY (CHICAGO, ILL.) 2024; 3:1020. [PMID: 38699565 PMCID: PMC11064876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 05/05/2024]
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative dementia worldwide. AD is a multifactorial disease that causes a progressive decline in memory and function precipitated by toxic beta-amyloid (Aβ) proteins, a key player in AD pathology. In 2022, 6.5 million Americans lived with AD, costing the nation $321billion. The standard of care for AD treatment includes acetylcholinesterase inhibitors (AchEIs), NMDA receptor antagonists, and monoclonal antibodies (mAbs). However, these methods are either: 1) ineffective in improving cognition, 2) unable to change disease progression, 3) limited in the number of therapeutic targets, 4) prone to cause severe side effects (brain swelling, microhemorrhages with mAb, and bradycardia and syncope with AchEIs), 5) unable to effectively cross the blood-brain barrier, and 6) lack of understanding of the aging process on the disease. mAbs are available to lower Aβ, but the difficulties of reducing the levels of the toxic Aβ proteins in the brain without triggering brain swelling or microhemorrhages associated with mAbs make the risk-benefit profile of mAbs unclear. A novel multitarget, effective, and safe non-invasive approach utilizing Repeated Electromagnetic Field Stimulation (REMFS) lowers Aβ levels in human neurons and memory areas, prevents neuronal death, stops disease progression, and improves memory without causing brain edema or bleeds in AD mice. This REMFS treatment has not been developed for humans because current EMF devices have poor penetration depth and inhomogeneous E-field distribution in the brain. Here, we discussed the biology of these effects in neurons and the design of optimal devices to treat AD.
Collapse
Affiliation(s)
- Felipe P Perez
- Department of Medicine, Indiana University School of Medicine, USA
| | - Jorge Morisaki
- Department of Bioengineering, University of Illinois at Chicago, Chicago, IL, USA
| | - Haitham Kanakri
- Department of Electrical and Computer Engineering, Indiana University-Purdue University, Indianapolis, IN, USA
| | - Maher Rizkalla
- Department of Electrical and Computer Engineering, Indiana University-Purdue University, Indianapolis, IN, USA
| |
Collapse
|
7
|
Triumbari EKA, Chiaravalloti A, Schillaci O, Mercuri NB, Liguori C. Positron Emission Tomography/Computed Tomography Imaging in Therapeutic Clinical Trials in Alzheimer's Disease: An Overview of the Current State of the Art of Research. J Alzheimers Dis 2024; 101:S603-S628. [PMID: 39422956 DOI: 10.3233/jad-240349] [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] [Indexed: 10/19/2024]
Abstract
The integration of positron emission tomography/computed tomography (PET/CT) has revolutionized the landscape of Alzheimer's disease (AD) research and therapeutic interventions. By combining structural and functional imaging, PET/CT provides a comprehensive understanding of disease pathology and response to treatment assessment. PET/CT, particularly with 2-deoxy-2-[fluorine-18]fluoro-D-glucose (18F-FDG), facilitates the visualization of glucose metabolism in the brain, enabling early diagnosis, staging, and monitoring of neurodegenerative disease progression. The advent of amyloid and tau PET imaging has further propelled the field forward, offering invaluable tools for tracking pathological hallmarks, assessing treatment response, and predicting clinical outcomes. While some therapeutic interventions targeting amyloid plaque load showed promising results with the reduction of cerebral amyloid accumulation over time, others failed to demonstrate a significant impact of anti-amyloid agents for reducing the amyloid plaques burden in AD brains. Tau PET imaging has conversely fueled the advent of disease-modifying therapeutic strategies in AD by supporting the assessment of neurofibrillary tangles of tau pathology deposition over time. Looking ahead, PET imaging holds immense promise for studying additional targets such as neuroinflammation, cholinergic deficit, and synaptic dysfunction. Advances in radiotracer development, dedicated brain PET/CT scanners, and Artificial Intelligence-powered software are poised to enhance the quality, sensitivity, and diagnostic power of molecular neuroimaging. Consequently, PET/CT remains at the forefront of AD research, offering unparalleled opportunities for unravelling the complexities of the disease and advancing therapeutic interventions, although it is not yet enough alone to allow patients' recruitment in therapeutic clinical trials.
Collapse
Affiliation(s)
| | - Agostino Chiaravalloti
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
- IRCCS Neuromed, Pozzilli, Italy
| | - Orazio Schillaci
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
| | - Nicola Biagio Mercuri
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
- Neurology Unit, University Hospital of Rome "Tor Vergata", Rome, Italy
| | - Claudio Liguori
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
- Neurology Unit, University Hospital of Rome "Tor Vergata", Rome, Italy
| |
Collapse
|
8
|
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
|
9
|
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
|
10
|
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
|
11
|
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
|
12
|
Fitzgerald GS, Chuchta TG, McNay EC. Insulin‐like growth factor‐2 is a promising candidate for the treatment and prevention of Alzheimer's disease. CNS Neurosci Ther 2023; 29:1449-1469. [PMID: 36971212 PMCID: PMC10173726 DOI: 10.1111/cns.14160] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 02/06/2023] [Accepted: 02/22/2023] [Indexed: 03/29/2023] Open
Abstract
Alzheimer's disease (AD) is the most common form of dementia. Current AD treatments slow the rate of cognitive decline, but do not restore lost function. One reason for the low efficacy of current treatments is that they fail to target neurotrophic processes, which are thought to be essential for functional recovery. Bolstering neurotrophic processes may also be a viable strategy for preventative treatment, since structural losses are thought to underlie cognitive decline in AD. The challenge of identifying presymptomatic patients who might benefit from preventative treatment means that any such treatment must meet a high standard of safety and tolerability. The neurotrophic peptide insulin-like growth factor-2 (IGF2) is a promising candidate for both treating and preventing AD-induced cognitive decline. Brain IGF2 expression declines in AD patients. In rodent models of AD, exogenous IGF2 modulates multiple aspects of AD pathology, resulting in (1) improved cognitive function; (2) stimulation of neurogenesis and synaptogenesis; and, (3) neuroprotection against cholinergic dysfunction and beta amyloid-induced neurotoxicity. Preclinical evidence suggests that IGF2 is likely to be safe and tolerable at therapeutic doses. In the preventative treatment context, the intranasal route of administration is likely to be the preferred method for achieving the therapeutic effect without risking adverse side effects. For patients already experiencing AD dementia, routes of administration that deliver IGF2 directly access the CNS may be necessary. Finally, we discuss several strategies for improving the translational validity of animal models used to study the therapeutic potential of IGF2.
Collapse
Affiliation(s)
| | | | - E C McNay
- University at Albany, Albany, New York, USA
| |
Collapse
|
13
|
Tani J, Yang YH, Chen CM, Siow CY, Chang TS, Yang K, Yao J, Hu CJ, Sung JY. Domain-Specific Cognitive Prosthesis for Face Memory and Recognition. Diagnostics (Basel) 2022; 12:diagnostics12092242. [PMID: 36140643 PMCID: PMC9497523 DOI: 10.3390/diagnostics12092242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 09/05/2022] [Accepted: 09/09/2022] [Indexed: 11/18/2022] Open
Abstract
The present study proposes a cognitive prosthesis device for face memory impairment as a proof-of-concept for the domain-specific cognitive prosthesis. Healthy subjects (n = 6) and a patient with poor face memory were enrolled. An acquaintance face recognition test with and without the use of cognitive prosthesis for face memory impairment, face recognition tests, quality of life, neuropsychological assessments, and machine learning performance of the cognitive prosthesis were followed-up throughout four weeks of real-world device use by the patient. The healthy subjects had an accuracy of 92.38 ± 4.41% and reaction time of 1.27 ± 0.12 s in the initial attempt of the acquaintance face recognition test, which changed to 80.48 ± 6.23% (p = 0.06) and 2.11 ± 0.20 s (p < 0.01) with prosthesis use. The patient had an accuracy of 74.29% and a reaction time of 6.65 s, which improved to 94.29% and 3.28 s with prosthesis use. After four weeks, the patient’s unassisted accuracy and reaction time improved to 100% and 1.23 s. Functional MRI study revealed activation of the left superior temporal lobe during face recognition task without prosthesis use and activation of the right precentral motor area with prosthesis use. The prosthesis could improve the patient’s performance by bypassing the brain area inefficient for facial recognition and employing the area more efficiently for the cognitive task.
Collapse
Affiliation(s)
- Jowy Tani
- Department of Neurology, Wan Fang Hospital, Taipei Medical University, Taipei 116079, Taiwan
- Biomed Innovation Center, Wan Fang Hospital, Taipei Medical University, Taipei 116079, Taiwan
- Department of Neurology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110301, Taiwan
- Taipei Medical University Biomed Accelerator, Taipei Medical University, Taipei 106339, Taiwan
- Taipei Medical University Biodesign Center, Taipei Medical University, Taipei 106339, Taiwan
- Taipei Neuroscience Institute, Taipei Medical University, New Taipei City 235041, Taiwan
| | - Yao-Hua Yang
- Biomed Innovation Center, Wan Fang Hospital, Taipei Medical University, Taipei 116079, Taiwan
| | - Chao-Min Chen
- Biomed Innovation Center, Wan Fang Hospital, Taipei Medical University, Taipei 116079, Taiwan
| | - Co Yih Siow
- Department of Physical Medicine and Rehabilitation, Taipei Medical University Hospital, Taipei 110301, Taiwan
| | - Tsui-San Chang
- Department of Neurology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110301, Taiwan
| | - Kai Yang
- MediXgraph Inc., Fremont, CA 94555, USA
| | - Jack Yao
- MediXgraph Inc., Fremont, CA 94555, USA
| | - Chaur-Jong Hu
- Department of Neurology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110301, Taiwan
- Taipei Neuroscience Institute, Taipei Medical University, New Taipei City 235041, Taiwan
- Department of Neurology, Shuang Ho Hospital, Taipei Medical University, New Taipei City 235041, Taiwan
- Correspondence: (C.-J.H.); (J.-Y.S.); Tel.: +886-2-2930-7930 (ext. 6940) (J.-Y.S.)
| | - Jia-Ying Sung
- Department of Neurology, Wan Fang Hospital, Taipei Medical University, Taipei 116079, Taiwan
- Department of Neurology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110301, Taiwan
- Taipei Neuroscience Institute, Taipei Medical University, New Taipei City 235041, Taiwan
- Correspondence: (C.-J.H.); (J.-Y.S.); Tel.: +886-2-2930-7930 (ext. 6940) (J.-Y.S.)
| |
Collapse
|
14
|
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
|
15
|
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
|
16
|
Treatment effects on event-related EEG potentials and oscillations in Alzheimer's disease. Int J Psychophysiol 2022; 177:179-201. [PMID: 35588964 DOI: 10.1016/j.ijpsycho.2022.05.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 05/11/2022] [Accepted: 05/12/2022] [Indexed: 11/23/2022]
Abstract
Alzheimer's disease dementia (ADD) is the most diffuse neurodegenerative disorder belonging to mild cognitive impairment (MCI) and dementia in old persons. This disease is provoked by an abnormal accumulation of amyloid-beta and tauopathy proteins in the brain. Very recently, the first disease-modifying drug has been licensed with reserve (i.e., Aducanumab). Therefore, there is a need to identify and use biomarkers probing the neurophysiological underpinnings of human cognitive functions to test the clinical efficacy of that drug. In this regard, event-related electroencephalographic potentials (ERPs) and oscillations (EROs) are promising candidates. Here, an Expert Panel from the Electrophysiology Professional Interest Area of the Alzheimer's Association and Global Brain Consortium reviewed the field literature on the effects of the most used symptomatic drug against ADD (i.e., Acetylcholinesterase inhibitors) on ERPs and EROs in ADD patients with MCI and dementia at the group level. The most convincing results were found in ADD patients. In those patients, Acetylcholinesterase inhibitors partially normalized ERP P300 peak latency and amplitude in oddball paradigms using visual stimuli. In these same paradigms, those drugs partially normalize ERO phase-locking at the theta band (4-7 Hz) and spectral coherence between electrode pairs at the gamma (around 40 Hz) band. These results are of great interest and may motivate multicentric, double-blind, randomized, and placebo-controlled clinical trials in MCI and ADD patients for final cross-validation.
Collapse
|
17
|
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
|
18
|
Martínez-Herrera M, Figueroa-Gerstenmaier S, López-Camacho PY, Millan-Pacheco C, Balderas-Altamirano MA, Mendoza-Franco G, García-Sierra F, Zavala-Ocampo LM, Basurto-Islas G. Multiadducts of C60 Modulate Amyloid-β Fibrillation with Dual Acetylcholinesterase Inhibition and Antioxidant Properties: In Vitro and In Silico Studies. J Alzheimers Dis 2022; 87:741-759. [DOI: 10.3233/jad-215412] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Background: Amyloid-β (Aβ) fibrils induce cognitive impairment and neuronal loss, leading to onset of Alzheimer’s disease (AD). The inhibition of Aβ aggregation has been proposed as a therapeutic strategy for AD. Pristine C60 has shown the capacity to interact with the Aβ peptide and interfere with fibril formation but induces significant toxic effects in vitro and in vivo. Objective: To evaluate the potential of a series of C60 multiadducts to inhibit the Aβ fibrillization. Methods: A series of C60 multiadducts with four to six diethyl malonyl and their corresponding disodium-malonyl substituents were synthesized as individual isomers. Their potential on Aβ fibrillization inhibition was evaluated in vitro, in cellulo, and silico. Antioxidant activity, acetylcholinesterase inhibition capacity, and toxicity were assessed in vitro. Results: The multiadducts modulate Aβ fibrils formation without inducing cell toxicity, and that the number and polarity of the substituents play a significant role in the adducts efficacy to modulate Aβ aggregation. The molecular mechanism of fullerene-Aβ interaction and modulation was identified. Furthermore, the fullerene derivatives exhibited antioxidant capacity and reduction of acetylcholinesterase activity. Conclusion: Multiadducts of C60 are novel multi-target-directed ligand molecules that could hold considerable promise as the starting point for the development of AD therapies.
Collapse
Affiliation(s)
- Melchor Martínez-Herrera
- Departamento de Ciencias Naturales e Ingeniería, Universidad Autónoma Metropolitana, Cuajimalpa, Ciudad de México, México
| | | | - Perla Y. López-Camacho
- Departamento de Ciencias Naturales e Ingeniería, Universidad Autónoma Metropolitana, Cuajimalpa, Ciudad de México, México
| | - Cesar Millan-Pacheco
- Facultad de Farmacia, Universidad Autónoma del Estado de Morelos, Cuernavaca, Morelos, México
| | | | - Graciela Mendoza-Franco
- Departamento de Biología Celular, Centro de Investigación y Estudios Avanzados del Instituto Politécnico Nacional, Ciudad de México, México
| | - Franciscos García-Sierra
- Departamento de Biología Celular, Centro de Investigación y Estudios Avanzados del Instituto Politécnico Nacional, Ciudad de México, México
| | - Lizeth M. Zavala-Ocampo
- Doctorado en Ciencias Biológicas y de la Salud, Universidad Autónoma Metropolitana, Cuajimalpa, Ciudad de México, México
| | - Gustavo Basurto-Islas
- División de Ciencias e Ingenierías, Universidad de Guanajuato, Campus León, León, Gto., Mexico
| |
Collapse
|
19
|
Andrade SM, Machado DGDS, Silva-Sauerc LD, Regis CT, Mendes CKTT, de Araújo JSS, de Araújo KDT, Costa LP, Queiroz MEBS, Leitão MM, Fernández-Calvo B. Effects of multisite anodal transcranial direct current stimulation combined with cognitive stimulation in patients with Alzheimer's disease and its neurophysiological correlates: A double-blind randomized clinical trial. Neurophysiol Clin 2022; 52:117-127. [PMID: 35339351 DOI: 10.1016/j.neucli.2022.02.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 02/25/2022] [Accepted: 02/28/2022] [Indexed: 01/02/2023] Open
Abstract
OBJECTIVES We aimed to examine the effects of multisite anodal transcranial direct current stimulation (tDCS) combined with cognitive stimulation (CS) over 2 months on cognitive performance and brain activity, and the relationship between them, in patients with Alzheimer's disease (AD). METHODS Patients with AD were randomly assigned to an active tDCS+CS (n=18) or a sham tDCS+CS (n=18) group. Cognitive performance was assessed using the Alzheimer Disease Assessment Scale-cognitive subscale (ADAS-cog) and brain activity using EEG (spectral power and coherence analysis) before and after the intervention. Multisite anodal tDCS (2 mA, 30 min) was applied over six brain regions [left and right dorsolateral prefrontal cortex (F3 and F4), Broca's area (F5), Wernicke's area (CP5), left and right somatosensory association cortex (P3 and P4)] for 24 sessions (three times a week). Both groups performed CS during tDCS. RESULTS Anodal tDCS+CS delays cognitive decline (ADAS-cog change) to a greater extent than sham tDCS+CS (-3.4±1.1 vs. -1.7±0.4; p=.03). Bilateral EEG coherence at high and low frequencies was greater for the active tDCS+CS than sham+CS group for most electrode pairs assessed (p < .05). The post-intervention ADAS-cog change score was predictive for EEG coherence at different sites (R²=.59 to .68; p < .05) in the active but not in the sham tDCS+CS group. CONCLUSION Anodal tDCS+CS improved overall cognitive function and changed EEG brain activity compared to sham tDCS+CS. Changes in cognitive performance were associated with changes in EEG measures of brain activity. Anodal tDCS+CS appears to be a promising therapeutic strategy to modulate cortical activity and improve cognitive function in patients with AD.
Collapse
Affiliation(s)
| | | | - Leandro da Silva-Sauerc
- Laboratory of Aging and Neurodegenerative Disorder, Department of Psychology, Federal University of Paraíba, João Pessoa, Brazil
| | - Cláudio Teixeira Regis
- Aging and Neuroscience Laboratory, Federal University of Paraíba, João Pessoa, PB, Brazil
| | | | | | | | - Larissa Pereira Costa
- Aging and Neuroscience Laboratory, Federal University of Paraíba, João Pessoa, PB, Brazil
| | | | | | - Bernardino Fernández-Calvo
- Laboratory of Aging and Neurodegenerative Disorder, Department of Psychology, Federal University of Paraíba, João Pessoa, Brazil; Department of Psychology, Faculty of Educational Sciences and Psychology, University of Córdoba, Córdoba, Spain; Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Córdoba, Spain
| |
Collapse
|
20
|
Dunn LB, Kim JP, Rostami M, Mondal S, Ryan K, Waraich A, Roberts LW, Palmer BW. Stakeholders' Perspectives regarding Participation in Neuromodulation-Based Dementia Intervention Research. J Empir Res Hum Res Ethics 2022; 17:29-38. [PMID: 34870511 PMCID: PMC9631956 DOI: 10.1177/15562646211060997] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
This study evaluated stakeholders' perspectives regarding participation in two hypothetical neuromodulation trials focused on individuals with Alzheimer's disease and related disorders (ADRDs). Stakeholders (i.e., individuals at risk for ADRDs [n = 56], individuals with experience as a caregiver for someone with a cognitive disorder [n = 60], and comparison respondents [n = 124]) were recruited via MTurk. Primary outcomes were willingness to enroll (or enroll one's loved one), feeling lucky to have the opportunity to enroll, and feeling obligated to enroll in two protocols (transcranial magnetic stimulation, TMS; deep brain stimulation, DBS). Relative to the Comparison group, the At Risk group endorsed higher levels of "feeling lucky" regarding both research protocols, and higher willingness to participate in the TMS protocol. These findings provide tentative reassurance regarding the nature of decision making regarding neurotechnology-based research on ADRDs. Further work is needed to evaluate the full range of potential influences on research participation.
Collapse
Affiliation(s)
- Laura B. Dunn
- Department of Psychiatry and Behavioral Sciences, Stanford University (USA)
| | - Jane P. Kim
- Department of Psychiatry and Behavioral Sciences, Stanford University (USA)
| | - Maryam Rostami
- Department of Psychiatry and Behavioral Sciences, Stanford University (USA)
| | - Sangeeta Mondal
- Department of Psychiatry and Behavioral Sciences, Stanford University (USA)
| | - Katie Ryan
- Department of Psychiatry and Behavioral Sciences, Stanford University (USA)
| | - Asees Waraich
- Keck School of Medicine, University of Southern California (USA)
| | | | - Barton W. Palmer
- Psychology Service, Veterans Affairs San Diego Healthcare System (USA)
- Department of Psychiatry, University of California, San Diego (USA)
| |
Collapse
|
21
|
Perez FP, Arvidson DM, Taylor TP, Rahmani M, Rizkalla M. Numerical Analysis and Design of an EMF Birdcage Wearable Device for the Treatment of Alzheimer’s Disease: A Feasibility Study. JOURNAL OF BIOMEDICAL SCIENCE AND ENGINEERING 2022; 15:219-227. [PMID: 36032690 PMCID: PMC9406889 DOI: 10.4236/jbise.2022.158020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
In this study, we performed a numerical analysis of a novel EMF Birdcage wearable device for the treatment of Alzheimer’s disease (AD). We designed the new device to generate and radiate a frequency of 64 MHz and a specific absorption rate (SAR) of 0.6 W/kg to a simulated human brain tissue. We determined these parameters from our experimental studies on primary human brain cultures at the Indiana University School of Medicine (IUSM). We found that this frequency and SAR decreased the toxic Aβ levels in the cell cultures. The frequency of 64 MHZ has good skin depth penetration, which will easily pass through the various head layers, including hair, skin, fat, dura, the cerebrospinal (CSF), and grey matter, and reach deeply into the brain tissues. The SAR of 0.6 W/kg was achieved with lower power input and energy, decreasing the probability of thermal injury. Therefore, these parameters enhance the safety of these potential treatments. This Birdcage device emulates a small-scale MRI machine, producing the same 64 MHz frequency at much lower operating input power. In this work, we utilized a high-frequency simulation system (HFSS/EMPro) software to produce the birdcage structure for the required EMF parameters. The 64 MHz radiating frequency produced the scattering S11 parameter of −15 dbs. We obtained a SAR of 0.6 W/kg when an input power of 100 W was applied. The coil dimensions were found to be near 15 cm in height and 22 cm in diameter, which fits in wearable systems. We found that the distribution of the electric field and SAR radiate homogeneously over the simulated human head with good penetration into the brain, which proves to be an appropriate potential therapeutic strategy for Alzheimer’s disease.
Collapse
Affiliation(s)
- Felipe P. Perez
- Department of Medicine, Division of General Internal Medicine and Geriatrics, Indiana University School of Medicine, Indianapolis, USA
| | - David Michael Arvidson
- Department of Electrical and Computer Engineering, Indiana University-Purdue University, Indianapolis, USA
| | - Tyler Phoenix Taylor
- Department of Electrical and Computer Engineering, Indiana University-Purdue University, Indianapolis, USA
| | - Maryam Rahmani
- Department of Electrical and Computer Engineering, Indiana University-Purdue University, Indianapolis, USA
| | - Maher Rizkalla
- Department of Electrical and Computer Engineering, Indiana University-Purdue University, Indianapolis, USA
| |
Collapse
|
22
|
Leblhuber F, Geisler S, Ehrlich D, Steiner K, Kurz K, Fuchs D. High Frequency Repetitive Transcranial Magnetic Stimulation Improves Cognitive Performance Parameters in Patients with Alzheimer's Disease - An Exploratory Pilot Study. Curr Alzheimer Res 2022; 19:681-688. [PMID: 36125835 DOI: 10.2174/1567205019666220920090919] [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: 04/04/2022] [Revised: 07/20/2022] [Accepted: 08/01/2022] [Indexed: 11/22/2022]
Abstract
BACKGROUND Currently available medication for Alzheimer's disease (AD) slows cognitive decline only temporarily but has failed to bring about long term positive effects. For this slowly progressive neurodegenerative disease, so far, no disease modifying therapy exists. OBJECTIVE The study aims to find out if non-pharmacologic non-invasive neuromodulatory repetitive transcranial magnetic stimulation (rTMS) may offer a new alternative or an add on therapeutic strategy against loss of cognitive functions. METHODS In this exploratory intervention study, safety and symptom development before and after frontopolar cortex stimulation (FPC) using intermittent theta burst stimulation (iTBS) at 10 subsequent working days was monitored as add-on treatment in 28 consecutive patients with AD. Out of these, 10 randomly selected patients received sham stimulation as a control. Serum concentrations of neurotransmitter precursor amino acids, immune activation and inflammation markers, brain-derived neurotrophic factor (BDNF), and nitrite were measured. RESULTS Treatment was well tolerated, and no serious adverse effects were observed. Improvement of cognition was detected by an increase in Mini Mental State Examination score (MMSE; p<0.01, paired rank test) and also by an increase in a modified repeat address phrase test, part of the 6-item cognitive impairment test (p<0.01). A trend to increase the clock drawing test (CDT; p = 0.08) was also found in the verum treated group. Furtheron, in 10 of the AD patients with additional symptoms of depression treated with iTBS, a significant decrease in the HAMD-7 scale (p<0.01) and a trend to lower serum phenylalanine concentrations (p = 0.08) was seen. No changes in the parameters tested were found in the sham treated patients. CONCLUSION Our preliminary results may indicate that iTBS is effective in the treatment of AD. Also a slight influence of iTBS on the metabolism of phenylalanine was found after 10 iTBS sessions. An impact of iTBS to influence the enzyme phenylalanine hydroxylase (PAH), as found in the previous series of treatment resistant depression, could not be seen in our first observational trial in 10 AD patients with comorbidity of depression. Longer treatment periods for several weeks in a higher number of AD patients with depression could cause more intense and disease modifying effects visible in different neurotransmitter concentrations important in the pathogenesis of AD.
Collapse
Affiliation(s)
| | - Simon Geisler
- Institute of Biological Chemistry, Biocenter, Innsbruck Medical University, Innsbruck, Austria
| | - Daniela Ehrlich
- Department of Gerontology, Kepler University Clinic, Linz, Austria
| | - Kostja Steiner
- Department of Gerontology, Kepler University Clinic, Linz, Austria
| | - Katharina Kurz
- Department of Internal Medicine, Innsbruck Medical University, Innsbruck, Austria
| | - Dietmar Fuchs
- Institute of Biological Chemistry, Biocenter, Innsbruck Medical University, Innsbruck, Austria
| |
Collapse
|
23
|
Azmi H. Neuromodulation for Cognitive Disorders: In Search of Lazarus? Neurol India 2021; 68:S288-S296. [PMID: 33318364 DOI: 10.4103/0028-3886.302469] [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: 11/04/2022]
Abstract
Alzheimer's disease (AD) and other forms of dementia can have a large impact on patients, their families, and for the society as a whole. Current medical treatments have not shown enough potential in treating or altering the course of the disease. Deep brain stimulation (DBS) has shown great neuromodulatory potential in Parkinson's disease, and there is a growing body of evidence for justifying its use in cognitive disorders. At the same time there is mounting interest at less invasive and alternative modes of neuromodulation for the treatment of AD. This manuscript is a brief review of the infrastructure of memory, the current understanding of the pathophysiology of AD, and the body of preclinical and clinical evidence for noninvasive and invasive neuromodulation modalities for the treatment of cognitive disorders and AD in particular.
Collapse
Affiliation(s)
- Hooman Azmi
- Department of Neurosurgery, Hackensack University Medical Center, Hackensack Meridian Health, Hackensack; New Jersey Brain and Spine Center, Oradell, New Jersey, USA
| |
Collapse
|
24
|
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: 451] [Impact Index Per Article: 112.8] [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
|
25
|
Perez FP, Maloney B, Chopra N, Morisaki JJ, Lahiri DK. Repeated electromagnetic field stimulation lowers amyloid-β peptide levels in primary human mixed brain tissue cultures. Sci Rep 2021; 11:621. [PMID: 33436686 PMCID: PMC7804462 DOI: 10.1038/s41598-020-77808-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Accepted: 11/17/2020] [Indexed: 02/07/2023] Open
Abstract
Late Onset Alzheimer's Disease is the most common cause of dementia, characterized by extracellular deposition of plaques primarily of amyloid-β (Aβ) peptide and tangles primarily of hyperphosphorylated tau protein. We present data to suggest a noninvasive strategy to decrease potentially toxic Aβ levels, using repeated electromagnetic field stimulation (REMFS) in primary human brain (PHB) cultures. We examined effects of REMFS on Aβ levels (Aβ40 and Aβ42, that are 40 or 42 amino acid residues in length, respectively) in PHB cultures at different frequencies, powers, and specific absorption rates (SAR). PHB cultures at day in vitro 7 (DIV7) treated with 64 MHz, and 1 hour daily for 14 days (DIV 21) had significantly reduced levels of secreted Aβ40 (p = 001) and Aβ42 (p = 0.029) peptides, compared to untreated cultures. PHB cultures (DIV7) treated at 64 MHz, for 1 or 2 hour during 14 days also produced significantly lower Aβ levels. PHB cultures (DIV28) treated with 64 MHz 1 hour/day during 4 or 8 days produced a similar significant reduction in Aβ40 levels. 0.4 W/kg was the minimum SAR required to produce a biological effect. Exposure did not result in cellular toxicity nor significant changes in secreted Aβ precursor protein-α (sAPPα) levels, suggesting the decrease in Aβ did not likely result from redirection toward the α-secretase pathway. EMF frequency and power used in our work is utilized in human magnetic resonance imaging (MRI, thus suggesting REMFS can be further developed in clinical settings to modulate Aβ deposition.
Collapse
Affiliation(s)
- Felipe P Perez
- Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Medicine, Division of General Internal Medicine and Geriatrics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Bryan Maloney
- Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Psychiatry, Institute of Psychiatric Research, Neuroscience Research Center, Indiana University School of Medicine, 320 W. 15th St, Indianapolis, IN, 46201, USA
| | - Nipun Chopra
- Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Psychiatry, Institute of Psychiatric Research, Neuroscience Research Center, Indiana University School of Medicine, 320 W. 15th St, Indianapolis, IN, 46201, USA
| | - Jorge J Morisaki
- Department of Bioengineering, University of Illinois at Chicago, Chicago, IL, 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, 320 W. 15th St, Indianapolis, IN, 46201, USA.
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA.
| |
Collapse
|
26
|
Toniolo S, Sen A, Husain M. Modulation of Brain Hyperexcitability: Potential New Therapeutic Approaches in Alzheimer's Disease. Int J Mol Sci 2020; 21:E9318. [PMID: 33297460 PMCID: PMC7730926 DOI: 10.3390/ijms21239318] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 11/30/2020] [Accepted: 12/05/2020] [Indexed: 12/12/2022] Open
Abstract
People with Alzheimer's disease (AD) have significantly higher rates of subclinical and overt epileptiform activity. In animal models, oligomeric Aβ amyloid is able to induce neuronal hyperexcitability even in the early phases of the disease. Such aberrant activity subsequently leads to downstream accumulation of toxic proteins, and ultimately to further neurodegeneration and neuronal silencing mediated by concomitant tau accumulation. Several neurotransmitters participate in the initial hyperexcitable state, with increased synaptic glutamatergic tone and decreased GABAergic inhibition. These changes appear to activate excitotoxic pathways and, ultimately, cause reduced long-term potentiation, increased long-term depression, and increased GABAergic inhibitory remodelling at the network level. Brain hyperexcitability has therefore been identified as a potential target for therapeutic interventions aimed at enhancing cognition, and, possibly, disease modification in the longer term. Clinical trials are ongoing to evaluate the potential efficacy in targeting hyperexcitability in AD, with levetiracetam showing some encouraging effects. Newer compounds and techniques, such as gene editing via viral vectors or brain stimulation, also show promise. Diagnostic challenges include identifying best biomarkers for measuring sub-clinical epileptiform discharges. Determining the timing of any intervention is critical and future trials will need to carefully stratify participants with respect to the phase of disease pathology.
Collapse
Affiliation(s)
- Sofia Toniolo
- Cognitive Neurology Group, Nuffield Department of Clinical Neurosciences, John Radcliffe Hospital, University of Oxford, Oxford OX3 9DU, UK;
- Wellcome Trust Centre for Integrative Neuroimaging, Department of Experimental Psychology, University of Oxford, Oxford OX2 6AE, UK
| | - Arjune Sen
- Oxford Epilepsy Research Group, Nuffield Department Clinical Neurosciences, John Radcliffe Hospital, Oxford OX3 9DU, UK;
| | - Masud Husain
- Cognitive Neurology Group, Nuffield Department of Clinical Neurosciences, John Radcliffe Hospital, University of Oxford, Oxford OX3 9DU, UK;
- Wellcome Trust Centre for Integrative Neuroimaging, Department of Experimental Psychology, University of Oxford, Oxford OX2 6AE, UK
| |
Collapse
|
27
|
Senbekov M, Saliev T, Bukeyeva Z, Almabayeva A, Zhanaliyeva M, Aitenova N, Toishibekov Y, Fakhradiyev I. The Recent Progress and Applications of Digital Technologies in Healthcare: A Review. Int J Telemed Appl 2020; 2020:8830200. [PMID: 33343657 PMCID: PMC7732404 DOI: 10.1155/2020/8830200] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Revised: 11/16/2020] [Accepted: 11/17/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND The implementation of medical digital technologies can provide better accessibility and flexibility of healthcare for the public. It encompasses the availability of open information on the health, treatment, complications, and recent progress on biomedical research. At present, even in low-income countries, diagnostic and medical services are becoming more accessible and available. However, many issues related to digital health technologies remain unmet, including the reliability, safety, testing, and ethical aspects. PURPOSE The aim of the review is to discuss and analyze the recent progress on the application of big data, artificial intelligence, telemedicine, block-chain platforms, smart devices in healthcare, and medical education. Basic Design. The publication search was carried out using Google Scholar, PubMed, Web of Sciences, Medline, Wiley Online Library, and CrossRef databases. The review highlights the applications of artificial intelligence, "big data," telemedicine and block-chain technologies, and smart devices (internet of things) for solving the real problems in healthcare and medical education. Major Findings. We identified 252 papers related to the digital health area. However, the number of papers discussed in the review was limited to 152 due to the exclusion criteria. The literature search demonstrated that digital health technologies became highly sought due to recent pandemics, including COVID-19. The disastrous dissemination of COVID-19 through all continents triggered the need for fast and effective solutions to localize, manage, and treat the viral infection. In this regard, the use of telemedicine and other e-health technologies might help to lessen the pressure on healthcare systems. Summary. Digital platforms can help optimize diagnosis, consulting, and treatment of patients. However, due to the lack of official regulations and recommendations, the stakeholders, including private and governmental organizations, are facing the problem with adequate validation and approbation of novel digital health technologies. In this regard, proper scientific research is required before a digital product is deployed for the healthcare sector.
Collapse
Affiliation(s)
- Maksut Senbekov
- S.D. Asfendiyarov Kazakh National Medical University, Almaty, Kazakhstan
| | - Timur Saliev
- S.D. Asfendiyarov Kazakh National Medical University, Almaty, Kazakhstan
| | | | | | | | - Nazym Aitenova
- NJSC “Astana Medical University”, Nur-Sultan, Kazakhstan
| | | | - Ildar Fakhradiyev
- S.D. Asfendiyarov Kazakh National Medical University, Almaty, Kazakhstan
| |
Collapse
|
28
|
Millan MJ, Dekeyne A, Gobert A, Brocco M, Mannoury la Cour C, Ortuno JC, Watson D, Fone KCF. Dual-acting agents for improving cognition and real-world function in Alzheimer's disease: Focus on 5-HT6 and D3 receptors as hubs. Neuropharmacology 2020; 177:108099. [PMID: 32525060 DOI: 10.1016/j.neuropharm.2020.108099] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 04/02/2020] [Accepted: 04/03/2020] [Indexed: 01/01/2023]
Abstract
To date, there are no interventions that impede the inexorable progression of Alzheimer's disease (AD), and currently-available drugs cholinesterase (AChE) inhibitors and the N-Methyl-d-Aspartate receptor antagonist, memantine, offer only modest symptomatic benefit. Moreover, a range of mechanistically-diverse agents (glutamatergic, histaminergic, monoaminergic, cholinergic) have disappointed in clinical trials, alone and/or in association with AChE inhibitors. This includes serotonin (5-HT) receptor-6 antagonists, despite compelling preclinical observations in rodents and primates suggesting a positive influence on cognition. The emphasis has so far been on high selectivity. However, for a multi-factorial disorder like idiopathic AD, 5-HT6 antagonists possessing additional pharmacological actions might be more effective, by analogy to "multi-target" antipsychotics. Based on this notion, drug discovery programmes have coupled 5-HT6 blockade to 5-HT4 agonism and inhibition of AchE. Further, combined 5-HT6/dopamine D3 receptor (D3) antagonists are of especial interest since D3 blockade mirrors 5-HT6 antagonism in exerting broad-based pro-cognitive properties in animals. Moreover, 5-HT6 and dopamine D3 antagonists promote neurocognition and social cognition via both distinctive and convergent actions expressed mainly in frontal cortex, including suppression of mTOR over-activation and reinforcement of cholinergic and glutamatergic transmission. In addition, 5-HT6 blockade affords potential anti-anxiety, anti-depressive and anti-epileptic properties, and antagonising 5-HT6 receptors may be associated with neuroprotective ("disease-modifying") properties. Finally D3 antagonism may counter psychotic episodes and D3 receptors themselves offer a promising hub for multi-target agents. The present article reviews the status of "R and D" into multi-target 5-HT6 and D3 ligands for improved treatment of AD and other neurodegenerative disorders of aging. This article is part of the special issue entitled 'Serotonin Research: Crossing Scales and Boundaries'.
Collapse
Affiliation(s)
- Mark J Millan
- Centre for Therapeutic Innovation in Neuropsychiatry, Institut de Recherche Servier, 78290, Croissy sur Seine, France.
| | - Anne Dekeyne
- Centre for Therapeutic Innovation in Neuropsychiatry, Institut de Recherche Servier, 78290, Croissy sur Seine, France
| | - Alain Gobert
- Centre for Therapeutic Innovation in Neuropsychiatry, Institut de Recherche Servier, 78290, Croissy sur Seine, France
| | - Mauricette Brocco
- Centre for Therapeutic Innovation in Neuropsychiatry, Institut de Recherche Servier, 78290, Croissy sur Seine, France
| | - Clotilde Mannoury la Cour
- Centre for Therapeutic Innovation in Neuropsychiatry, Institut de Recherche Servier, 78290, Croissy sur Seine, France
| | - Jean-Claude Ortuno
- Centre for Excellence in Chemistry, Institut de Recherche Servier, 78290, Croissy sur Seine, France
| | - David Watson
- School of Life Sciences, Queen's Medical Centre, The University of Nottingham, NG7 2UH, England, UK
| | - Kevin C F Fone
- School of Life Sciences, Queen's Medical Centre, The University of Nottingham, NG7 2UH, England, UK
| |
Collapse
|
29
|
Jiang Y, Yang H, Zhao J, Wu Y, Zhou X, Cheng Z. Reliability and concurrent validity of Alzheimer's disease assessment scale - Cognitive subscale, Chinese version (ADAS-Cog-C) among Chinese community-dwelling older people population. Clin Neuropsychol 2020; 34:43-53. [PMID: 32279575 DOI: 10.1080/13854046.2020.1750704] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
OBJECTIVE To evaluate reliability and concurrent validity of the Alzheimer's Disease Assessment Scale - Cognitive Subscale, Chinese Version (ADAS-Cog-C) among Chinese community older adults. METHOD Three groups, comprising of 1,276 community-dwelling older adults, were included in this study: a normal control (NC), a mild cognitive impairment (MCI), and an Alzheimer's disease (AD) group. All participants were assessed through ADAS-Cog-C, clinical interviews, physical examinations, Mini Mental State Examination (MMSE), and the Clinical Dementia Rating Scale (CDR). Internal consistency was assessed to evaluate the reliability of ADAS-Cog-C. Pearson and Spearman correlation coefficients were calculated to evaluate the concurrent validity between ADAS-Cog-C, MMSE, and CDR. RESULTS Overall, the Cronbach's alpha coefficients of ADAS-Cog-C for the AD and MCI groups were 0.843 and 0.554, respectively. The split-half reliability coefficients for the AD and MCI groups were 0.860 and 0.539, respectively. ADAS-Cog-C scores were negatively correlated with MMSE scores (r = -0.706, p < 0.001) and positively associated with CDR scores (r = 0.546, p < 0.001). After excluding the MCI group from the analysis, the internal consistency of ADAS-Cog-C for the total population improved (α = 0.813, r hh = 0.852, all p < 0.001), as did the correlation between ADAS-Cog-C and MMSE (r = -0.828, p < 0.001) and CDR (r = 0.429, all p < 0.001) scores. CONCLUSIONS ADAS-Cog-C has good internal consistency and concurrent validity for assessing Chinese community older adults with AD, but poor consistency, good concurrent validity with the MMSE while moderate concurrent validity with the CDR for MCI.
Collapse
Affiliation(s)
- Yan Jiang
- Department of Geriatric Psychiatry, The affiliated Wuxi Mental Health Center of Nanjing Medical University, Wuxi, China
| | - Hongyu Yang
- Department of Geriatric Psychiatry, The affiliated Wuxi Mental Health Center of Nanjing Medical University, Wuxi, China
| | - Jinfa Zhao
- Department of Psychology, Graduate School of Wannan Medical College, Wuhu, China
| | - Yue Wu
- Department of Geriatric Psychiatry, The affiliated Wuxi Mental Health Center of Nanjing Medical University, Wuxi, China
| | - Xiaoqin Zhou
- Department of Geriatric Psychiatry, The affiliated Wuxi Mental Health Center of Nanjing Medical University, Wuxi, China
| | - Zaohuo Cheng
- Department of Geriatric Psychiatry, The affiliated Wuxi Mental Health Center of Nanjing Medical University, Wuxi, China
| |
Collapse
|