1
|
Isaković J, Chin BD, Oberwinter M, Rance HK. From lab coats to clinical trials: Evolution and application of electromagnetic fields for ischemic stroke rehabilitation and monitoring. Brain Res 2025; 1850:149391. [PMID: 39662791 DOI: 10.1016/j.brainres.2024.149391] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 12/05/2024] [Accepted: 12/06/2024] [Indexed: 12/13/2024]
Abstract
Stroke is a neurovascular disorder which stands as one of the leading causes of death and disability worldwide, resulting in motor and cognitive impairment. Although the treatment approach depends on the time elapsed, the type of stroke and the availability of care centers, common interventions include thrombectomy or the administration of a tissue plasminogen activator (tPA). While these methods restore blood flow, they fall short in helping patients regain lost function. With that, recent years have seen a rise in novel methods, one of which is the use of electromagnetic fields (EMFs). Due to their ability to impact the charges in their vicinity, thereby altering the immune response and cell signaling, EMFs became suitable candidates for stroke rehabilitation. Based on their characteristics, therapeutic EMFs can be categorized into transcranial magnetic stimulation (TMS), transcranial direct current stimulation (tDCS), pulsed (PEMFs) and low frequency (LF-EMFs) electromagnetic fields, among others. In addition to treatment, EMFs are being explored for stroke monitoring, utilizing external EMFs for imaging or recording innate EMFs linked to neural activity. Drawing from research on the effects of EMFs, this review aims to provide a comprehensive overview of the physical principles and molecular mechanisms underlying the action of EMFs, along with a discussion of their application in preclinical studies and clinical trials. Finally, this paper not only addresses the importance of treatment availability and potential side-effects, but also delves into the technical and ethical challenges associated with the use of EMFs, while exploring their prospects and future opportunities.
Collapse
Affiliation(s)
- Jasmina Isaković
- School of Medicine, European University Cyprus - Frankfurt Branch, 60488 Frankfurt am Main, Germany.
| | - Benjamin Daniel Chin
- School of Medicine, European University Cyprus - Frankfurt Branch, 60488 Frankfurt am Main, Germany
| | - Moritz Oberwinter
- School of Medicine, European University Cyprus - Frankfurt Branch, 60488 Frankfurt am Main, Germany
| | - Hannah Katarina Rance
- School of Medicine, European University Cyprus - Frankfurt Branch, 60488 Frankfurt am Main, Germany
| |
Collapse
|
2
|
Brown JC, Kweon J, Sharma P, Siddiqi SH, Isserles M, Ressler KJ. Critically Assessing the Unanswered Questions of How, Where, and When to Induce Plasticity in the Posttraumatic Stress Disorder Network With Transcranial Magnetic Stimulation. Biol Psychiatry 2025; 97:392-404. [PMID: 38909668 DOI: 10.1016/j.biopsych.2024.06.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2023] [Revised: 06/02/2024] [Accepted: 06/10/2024] [Indexed: 06/25/2024]
Abstract
Extinction of traumatic memory, a primary treatment approach (termed exposure therapy) in posttraumatic stress disorder (PTSD), occurs through relearning and may be subserved at the molecular level by long-term potentiation of relevant circuits. In parallel, repetitive transcranial magnetic stimulation (TMS) is thought to work through long-term potentiation-like mechanisms and may provide a novel, safe, and effective treatment for PTSD. In a recent failed randomized controlled trial we emphasized the necessity of correctly identifying cortical targets, the directionality of TMS protocols, and the role of memory activation. Here, we provide a systematic review of TMS for PTSD to further identify how, where, and when TMS treatment should be delivered to alleviate PTSD symptoms. We conducted a systematic review of the literature by searching for repetitive TMS clinical trials involving patients with PTSD and outcomes. We searched MEDLINE through October 25, 2023, for "TMS and PTSD" and "transcranial magnetic stimulation and posttraumatic stress disorder." Thirty-one publications met our inclusion criteria (k = 17 randomized controlled trials, k = 14 open label). Randomized controlled trial protocols were varied in terms of TMS protocols, cortical TMS targets, and memory activation protocols. There was no clear superiority of low-frequency (k = 5) versus high-frequency (k = 6) protocols or by stimulation location. Memory provocation or exposure protocols (k = 7) appear to enhance response. Overall, TMS appears to be effective in treating PTSD symptoms across a variety of TMS frequencies, hemispheric target differences, and exposure protocols. Disparate protocols may be conceptually harmonized when viewed as potentiating proposed anxiolytic networks or suppressing anxiogenic networks.
Collapse
Affiliation(s)
- Joshua C Brown
- Division of Depression and Anxiety Disorders, McLean Hospital, Belmont, Massachusetts; Department of Psychiatry, Harvard Medical School, Boston, Massachusetts.
| | - Jamie Kweon
- Division of Depression and Anxiety Disorders, McLean Hospital, Belmont, Massachusetts
| | - Prayushi Sharma
- Division of Depression and Anxiety Disorders, McLean Hospital, Belmont, Massachusetts
| | - Shan H Siddiqi
- Department of Psychiatry, Harvard Medical School, Boston, Massachusetts; Center for Brain Circuit Therapeutics, Brigham and Women's Hospital, Boston, Massachusetts
| | - Moshe Isserles
- Department of Psychiatry, Hadassah Hebrew University Medical Center, Jerusalem, Israel
| | - Kerry J Ressler
- Division of Depression and Anxiety Disorders, McLean Hospital, Belmont, Massachusetts; Department of Psychiatry, Harvard Medical School, Boston, Massachusetts.
| |
Collapse
|
3
|
Hanauske T, Koretz CC, Jungenitz T, Roeper J, Drakew A, Deller T. Electrophysiologically calibrated optogenetic stimulation of dentate granule cells mitigates dendritic spine loss in denervated organotypic entorhino-hippocampal slice cultures. Sci Rep 2025; 15:4563. [PMID: 39915664 PMCID: PMC11802742 DOI: 10.1038/s41598-025-88536-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Accepted: 01/29/2025] [Indexed: 02/09/2025] Open
Abstract
Organotypic slice cultures (OTCs) are versatile tools for studying long-term structure-function relationships of neurons within a defined network (e.g. hippocampus). We developed a method for repeated experimenter-controlled activation of hippocampal granule cells (GCs) in OTCs within the incubator. After several days of contact-free photonic stimulation, we were able to ameliorate entorhinal denervation-induced structural damage in GCs. To achieve this outcome, we had to calibrate the intensity and duration of optogenetic (light) pulses using whole-cell electrophysiological recordings and multi-cell calcium imaging. Our findings showed that ChR2-expressing cells generated action potentials (APs) or calcium transients in response to illumination but were otherwise functionally indistinguishable from non-transduced GCs within the same neural circuit. However, the threshold for AP firing in single GCs varied based on the stimulus light intensity and the expression levels of ChR2. This information allowed us to calibrate light intensity for chronic stimulations. Denervated GCs exhibited significant spine loss four days post-denervation, but this detrimental effect was mitigated when AP firing was induced at a physiological GC bursting rate. Phototoxic damage caused by chronic light exposure was significantly reduced if illuminated with longer wavelength and by adding antioxidants to the culture medium. Our study presents a versatile approach for concurrent non-invasive manipulation and observation of neural circuit activity and remodeling in vitro.
Collapse
Affiliation(s)
- Tijana Hanauske
- Institute for Clinical Neuroanatomy, Dr. Senckenberg Anatomy, Faculty of Medicine, Goethe University Frankfurt, Theodor-Stern-Kai 7, 60590, Frankfurt, Germany.
| | - Carolin Christina Koretz
- Institute for Clinical Neuroanatomy, Dr. Senckenberg Anatomy, Faculty of Medicine, Goethe University Frankfurt, Theodor-Stern-Kai 7, 60590, Frankfurt, Germany
- Institute for Neurophysiology, Faculty of Medicine, Goethe University Frankfurt, Theodor-Stern-Kai 7, 60590, Frankfurt, Germany
| | - Tassilo Jungenitz
- Institute for Clinical Neuroanatomy, Dr. Senckenberg Anatomy, Faculty of Medicine, Goethe University Frankfurt, Theodor-Stern-Kai 7, 60590, Frankfurt, Germany
| | - Jochen Roeper
- Institute for Neurophysiology, Faculty of Medicine, Goethe University Frankfurt, Theodor-Stern-Kai 7, 60590, Frankfurt, Germany
| | - Alexander Drakew
- Institute for Clinical Neuroanatomy, Dr. Senckenberg Anatomy, Faculty of Medicine, Goethe University Frankfurt, Theodor-Stern-Kai 7, 60590, Frankfurt, Germany
| | - Thomas Deller
- Institute for Clinical Neuroanatomy, Dr. Senckenberg Anatomy, Faculty of Medicine, Goethe University Frankfurt, Theodor-Stern-Kai 7, 60590, Frankfurt, Germany
| |
Collapse
|
4
|
Liao C, Dua AN, Wojtasiewicz C, Liston C, Kwan AC. Structural neural plasticity evoked by rapid-acting antidepressant interventions. Nat Rev Neurosci 2025; 26:101-114. [PMID: 39558048 DOI: 10.1038/s41583-024-00876-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/17/2024] [Indexed: 11/20/2024]
Abstract
A feature in the pathophysiology of major depressive disorder (MDD), a mood disorder, is the impairment of excitatory synapses in the prefrontal cortex. Intriguingly, different types of treatment with fairly rapid antidepressant effects (within days or a few weeks), such as ketamine, electroconvulsive therapy and non-invasive neurostimulation, seem to converge on enhancement of neural plasticity. However, the forms and mechanisms of plasticity that link antidepressant interventions to the restoration of excitatory synaptic function are still unknown. In this Review, we highlight preclinical research from the past 15 years showing that ketamine and psychedelic drugs can trigger the growth of dendritic spines in cortical pyramidal neurons. We compare the longitudinal effects of various psychoactive drugs on neuronal rewiring, and we highlight rapid onset and sustained time course as notable characteristics for putative rapid-acting antidepressant drugs. Furthermore, we consider gaps in the current understanding of drug-evoked in vivo structural plasticity. We also discuss the prospects of using synaptic remodelling to understand other antidepressant interventions, such as repetitive transcranial magnetic stimulation. Finally, we conclude that structural neural plasticity can provide unique insights into the neurobiological actions of psychoactive drugs and antidepressant interventions.
Collapse
Affiliation(s)
- Clara Liao
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
- Interdepartmental Neuroscience Program, Yale University School of Medicine, New Haven, CT, USA
| | - Alisha N Dua
- Department of Psychiatry, Weill Cornell Medicine, New York, NY, USA
| | | | - Conor Liston
- Department of Psychiatry, Weill Cornell Medicine, New York, NY, USA
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Alex C Kwan
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA.
- Department of Psychiatry, Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|
5
|
Ji Y, Yang C, Pang X, Yan Y, Wu Y, Geng Z, Hu W, Hu P, Wu X, Wang K. Repetitive transcranial magnetic stimulation in Alzheimer's disease: effects on neural and synaptic rehabilitation. Neural Regen Res 2025; 20:326-342. [PMID: 38819037 PMCID: PMC11317939 DOI: 10.4103/nrr.nrr-d-23-01201] [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: 07/19/2023] [Revised: 10/23/2023] [Accepted: 12/13/2023] [Indexed: 06/01/2024] Open
Abstract
Alzheimer's disease is a neurodegenerative disease resulting from deficits in synaptic transmission and homeostasis. The Alzheimer's disease brain tends to be hyperexcitable and hypersynchronized, thereby causing neurodegeneration and ultimately disrupting the operational abilities in daily life, leaving patients incapacitated. Repetitive transcranial magnetic stimulation is a cost-effective, neuro-modulatory technique used for multiple neurological conditions. Over the past two decades, it has been widely used to predict cognitive decline; identify pathophysiological markers; promote neuroplasticity; and assess brain excitability, plasticity, and connectivity. It has also been applied to patients with dementia, because it can yield facilitatory effects on cognition and promote brain recovery after a neurological insult. However, its therapeutic effectiveness at the molecular and synaptic levels has not been elucidated because of a limited number of studies. This study aimed to characterize the neurobiological changes following repetitive transcranial magnetic stimulation treatment, evaluate its effects on synaptic plasticity, and identify the associated mechanisms. This review essentially focuses on changes in the pathology, amyloidogenesis, and clearance pathways, given that amyloid deposition is a major hypothesis in the pathogenesis of Alzheimer's disease. Apoptotic mechanisms associated with repetitive transcranial magnetic stimulation procedures and different pathways mediating gene transcription, which are closely related to the neural regeneration process, are also highlighted. Finally, we discuss the outcomes of animal studies in which neuroplasticity is modulated and assessed at the structural and functional levels by using repetitive transcranial magnetic stimulation, with the aim to highlight future directions for better clinical translations.
Collapse
Affiliation(s)
- Yi Ji
- Department of Neurology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui Province, China
- Anhui Province Key Laboratory of Cognition and Neuropsychiatric Disorders, Hefei, Anhui Province, China
| | - Chaoyi Yang
- Department of Neurology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui Province, China
- Anhui Province Key Laboratory of Cognition and Neuropsychiatric Disorders, Hefei, Anhui Province, China
| | - Xuerui Pang
- Department of Neurology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui Province, China
- Anhui Province Key Laboratory of Cognition and Neuropsychiatric Disorders, Hefei, Anhui Province, China
| | - Yibing Yan
- Department of Neurology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui Province, China
- Anhui Province Key Laboratory of Cognition and Neuropsychiatric Disorders, Hefei, Anhui Province, China
| | - Yue Wu
- Department of Neurology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui Province, China
| | - Zhi Geng
- Department of Neurology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui Province, China
- Anhui Province Key Laboratory of Cognition and Neuropsychiatric Disorders, Hefei, Anhui Province, China
| | - Wenjie Hu
- Department of Neurology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui Province, China
- Anhui Province Key Laboratory of Cognition and Neuropsychiatric Disorders, Hefei, Anhui Province, China
| | - Panpan Hu
- Department of Neurology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui Province, China
- Anhui Province Key Laboratory of Cognition and Neuropsychiatric Disorders, Hefei, Anhui Province, China
- Collaborative Innovation Center of Neuropsychiatric Disorders and Mental Health, Hefei, Anhui Province, China
| | - Xingqi Wu
- Department of Neurology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui Province, China
- Anhui Province Key Laboratory of Cognition and Neuropsychiatric Disorders, Hefei, Anhui Province, China
- Collaborative Innovation Center of Neuropsychiatric Disorders and Mental Health, Hefei, Anhui Province, China
| | - Kai Wang
- Department of Neurology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui Province, China
- Anhui Province Key Laboratory of Cognition and Neuropsychiatric Disorders, Hefei, Anhui Province, China
- Institute of Artificial Intelligence, Hefei Comprehensive National Science Center, Hefei, Anhui Province, China
- Department of Psychology and Sleep Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui Province, China
| |
Collapse
|
6
|
Fan S, Wang W, Zheng X. Repetitive Transcranial Magnetic Stimulation for the Treatment of Spinal Cord Injury: Current Status and Perspective. Int J Mol Sci 2025; 26:825. [PMID: 39859537 PMCID: PMC11766194 DOI: 10.3390/ijms26020825] [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: 12/26/2024] [Revised: 01/13/2025] [Accepted: 01/17/2025] [Indexed: 01/27/2025] Open
Abstract
Spinal cord injury (SCI) can lead to devastating dysfunctions and complications, significantly impacting patients' quality of life and aggravating the burden of disease. Since the main pathological mechanism of SCI is the disruption of neuronal circuits, the primary therapeutic strategy for SCI involves reconstructing and activating circuits to restore neural signal transmission. Repetitive transcranial magnetic stimulation (rTMS), a noninvasive brain stimulation technique, can modulate the function or state of the nervous system by pulsed magnetic fields. Here, we discuss the basic principles and potential mechanisms of rTMS for treating SCI, including promoting the reconstruction of damaged circuits in the spinal cord, activating reorganization of the cerebral cortex and circuits, modulating the balance of inputs to motoneurons, improving the microenvironment and intrinsic regeneration ability in SCI. Based on these mechanisms, we provide an overview of the therapeutic effects of rTMS in SCI patients with motor dysfunction, spasticity and neuropathic pain. We also discuss the challenges and prospectives of rTMS, especially the potential of combination therapy of rTMS and neural progenitor cell transplantation, and the synergistic effects on promoting regeneration, relay formation and functional connectivity. This review is expected to offer a relatively comprehensive understanding and new perspectives of rTMS for SCI treatment.
Collapse
Affiliation(s)
- Shu Fan
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China;
- Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Wei Wang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China;
- Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan 430030, China
- Key Laboratory of Neurological Diseases of Chinese Ministry of Education, the School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Xiaolong Zheng
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China;
- Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan 430030, China
| |
Collapse
|
7
|
Ong RCS, Tang AD. Subthreshold repetitive transcranial magnetic stimulation induces cortical layer-, brain region-, and protocol-dependent neural plasticity. SCIENCE ADVANCES 2025; 11:eado6705. [PMID: 39772671 PMCID: PMC11708880 DOI: 10.1126/sciadv.ado6705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/17/2024] [Accepted: 12/02/2024] [Indexed: 01/11/2025]
Abstract
Repetitive transcranial magnetic stimulation (rTMS) is commonly used to study the brain or as a treatment for neurological disorders, but the neural circuits and molecular mechanisms it affects remain unclear. To determine the molecular mechanisms of rTMS and the brain regions they occur in, we used spatial transcriptomics to map changes to gene expression across the mouse brain in response to two commonly used rTMS protocols. Our results revealed that rTMS alters the expression of genes related to several cellular processes and neural plasticity mechanisms across the brain, which was both brain region- and rTMS protocol-dependent. In the cortex, the effect of rTMS was dependent not only on the cortical region but also on each cortical layer. These findings uncover the diverse molecular mechanisms induced by rTMS, which will be useful in interpreting its effects on cortical and subcortical circuits.
Collapse
Affiliation(s)
- Rebecca C. S. Ong
- Experimental and Regenerative Neurosciences, The University of Western Australia, Perth, Australia
- Perron Institute for Neurological and Translational Sciences, Perth, Australia
| | - Alexander D. Tang
- Experimental and Regenerative Neurosciences, The University of Western Australia, Perth, Australia
- Perron Institute for Neurological and Translational Sciences, Perth, Australia
- Pharmacology and Toxicology Discipline, School of Biomedical Sciences, The University of Western Australia, Perth, Australia
| |
Collapse
|
8
|
Xing XX, Wu JJ, Qu J, Ma J, Xu R, Zhu Y, Zheng MX, Hua XY, Xu JG. Rewiring the disordered connectome with circuit-based paired stimulation after stroke-a randomized, double-blind and controlled Phase II trial. Brain Commun 2024; 6:fcae437. [PMID: 39697832 PMCID: PMC11653076 DOI: 10.1093/braincomms/fcae437] [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: 07/16/2023] [Revised: 07/15/2024] [Accepted: 12/04/2024] [Indexed: 12/20/2024] Open
Abstract
The cortico-cortical paired associative stimulation, a combined stimulation based on two brain regions, may be an effective strategy for stroke rehabilitation. Our aim was to confirm that the cortico-cortical paired associative stimulation strengthens the connection between brain regions in the motor circuit and promotes improvements in motor function. This was a randomized double-blind, controlled Phase II trial. 44 Stroke patients were treated in a rehabilitation hospital from October 2020 to January 2021 and were randomly assigned to the sham stimulation group and the cortico-cortical paired associative stimulation group. Patients in both groups received 12 days of rehabilitation therapy. Cortico-cortical paired associative stimulation group received one treatment of cortico-cortical paired associative stimulation invention. Both groups received behavioural assessments such as the Fugl-Meyer upper-extremity scale and resting-state functional MRI scans prior to the intervention and on Day 14. 40 patients completed the intervention session. The results of Fugl-Meyer upper-extremity scale showed a more significant improvement in motor function in the cortico-cortical paired associative stimulation group (6.33 ± 1.29) than in the sham stimulation group (3.16 ± 1.38) (P < 0.001). The functional connectivity showed that cortico-cortical paired associative stimulation strengthens connections between brain regions. Correlation analysis confirmed that the enhancement of functional connectivity was positively correlated with the recovery of Fugl-Meyer upper-extremity scale (r2 = 0.146, P = 0.034; r2 = 0.211, P = 0.0093). The results of functional connectivity suggest that cortico-cortical paired associative stimulation strengthens connections between brain regions. It is expected that this study will provide a positive viewpoint for the neurorehabilitation of stroke patients based on the circuit-level plasticity. (Chinese Clinical Trial Registry: ChiCTR2000036685).
Collapse
Affiliation(s)
- Xiang-Xin Xing
- Rehabilitation Center, Qilu Hospital of Shandong University, Qilu Hospital of Shandong University, Jinan 250012, China
- Department of Rehabilitation Medicine, Yueyang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, China
- Engineering Research Center of Traditional Chinese Medicine Intelligent Rehabilitation, Ministry of Education, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Jia-Jia Wu
- Department of Rehabilitation Medicine, Yueyang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, China
- Engineering Research Center of Traditional Chinese Medicine Intelligent Rehabilitation, Ministry of Education, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Jiao Qu
- Shanghai Songjiang District Central Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 201600, China
| | - Jie Ma
- Department of Rehabilitation Medicine, Yueyang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, China
- Engineering Research Center of Traditional Chinese Medicine Intelligent Rehabilitation, Ministry of Education, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Rong Xu
- YangZhi Rehabilitation Hospital, TongJi University, Shanghai 201600, China
| | - Yu Zhu
- Department of Physical Medicine and Rehabilitation, State University of New York Upstate Medical University, Syracuse 13290, USA
| | - Mou-Xiong Zheng
- Engineering Research Center of Traditional Chinese Medicine Intelligent Rehabilitation, Ministry of Education, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
- Department of Orthopedics, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Xu-Yun Hua
- Engineering Research Center of Traditional Chinese Medicine Intelligent Rehabilitation, Ministry of Education, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
- Department of Orthopedics, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Jian-Guang Xu
- Engineering Research Center of Traditional Chinese Medicine Intelligent Rehabilitation, Ministry of Education, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
- School of Rehabilitation Science, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| |
Collapse
|
9
|
Espinosa Mendoza TA, Oviedo Lara AR, Henk Jordan G, Sampieri-Cabrera R, Perez Martinez LE. Effects of Low-Intensity Transcranial Magnetic Stimulation in Neuropsychological Development of Pediatric Subjects With Autism Spectrum Disorder: A Longitudinal Retrospective Approach. Cureus 2024; 16:e76569. [PMID: 39877774 PMCID: PMC11774318 DOI: 10.7759/cureus.76569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/29/2024] [Indexed: 01/31/2025] Open
Abstract
BACKGROUND Autism spectrum disorder (ASD) is a heterogeneous neurobiological condition characterized by behavioral problems and delayed neurodevelopment. Although transcranial magnetic stimulation (TMS) has been proposed as an alternative treatment for patients with ASD because of its promising benefits in reducing repetitive behaviors and enhancing executive functions, the use of high-intensity pulses (Hi-TMS) appears to be related to the side effects of the therapy. Low-intensity TMS (Li-TMS) has been partially investigated, but it may have clinical effects on ASD and simultaneously increase treatment safety. METHODS In this study, the effects of combined intervention with Li-TMS and conventional therapies were evaluated in 35 patients from Ecuador (six female and 29 male), aged between three and seven years, with a confirmed diagnosis of ASD. Each subject received conventional therapies twice a week (for four weeks) provided at the research center (psychological, occupational, speech, and neuro-psychomotor therapies) alongside daily Li-TMS sessions at 1 Hz and 9 mT of intensity targeting the left dorsolateral prefrontal cortex (L-DLPFC) for 45 min for four weeks (16 sessions in total). The Battelle Developmental Inventory (BDI), first edition, was applied before and after Li-TMS therapy to evaluate its clinical effects in subjects with ASD. Weekly follow-up assessments and parent questionnaires were administered to identify any adverse events. RESULTS In all BDI domains, a significant statistical difference was observed between the pre- and post-intervention averages, supported by extremely low p-values (less than 0.001 in all cases). The personal, social, motor, cognitive, and communicative skills of all the study participants increased after Li-TMS therapy. At the same time, the calculated maturational delay had a significant decrease, suggesting an improvement of ~7.78 months in the neurodevelopment of the ASD subjects. However, age was also found to be a possible cause for these changes in development and maturation. No adverse effects were observed. CONCLUSIONS Both variants of TMS, Hi-TMS and Li-TMS, have proven to be promising treatments for subjects with ASD, improving social and cognitive abilities. This investigation suggests that the combination of conventional therapies and 16 sessions of Li-TMS as a treatment for individuals with ASD had significant clinical progress, specifically in maturation development according to BDI. In addition, the use of low-intensity magnetic fields may allow for safer pulse delivery in pediatric subjects, as no side effects were reported in this study.
Collapse
Affiliation(s)
| | | | - Gabriel Henk Jordan
- Neurosciences, Centro de Especialidades Neuropsicologicas Neuroinnova, Guayaquil, ECU
| | - Raúl Sampieri-Cabrera
- Foresight, Centro de Ciencias de la Complejidad, Universidad Nacional Autónoma de México, Mexico, MEX
- Department of Physiology, Facultad de Medicina, Universidad Nacional Autónoma de México, México, MEX
| | | |
Collapse
|
10
|
Kweon J, Fukuda AM, Gobin AP, Haq L, Carpenter LL, Brown JC. Effect of sleep quality on repetitive transcranial magnetic stimulation outcomes in depression. Front Psychiatry 2024; 15:1458696. [PMID: 39376965 PMCID: PMC11456523 DOI: 10.3389/fpsyt.2024.1458696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Accepted: 09/04/2024] [Indexed: 10/09/2024] Open
Abstract
Introduction While repetitive transcranial magnetic stimulation (rTMS) is effective for 50-60% of those treatment-resistant depression, it is critical to identify predictors of response for optimal patient selection to improve therapy. Insomnia is a known symptom of depression that is both correlated with depression severity and associated with poor antidepressant response. Therefore, understanding this relationship may open new opportunities for the optimization of rTMS treatment. We aimed to explore whether baseline sleep quality, specifically insomnia, is associated with rTMS outcomes in a naturalistic sample of 975 patients (age 18-90; 63.9% F) receiving a standard course of rTMS treatment from two outpatient TMS clinics located within psychiatric hospitals in the United States. One site additionally collected information on concurrent medication use on 350 patients; among these, we examined whether pharmacological treatment of insomnia affected TMS treatment response. Methods Depression was measured using the 30-item Inventory of Depressive Symptomology Self Report (IDS-SR) in site one and an abbreviated 16-item Quick Inventory of Depressive Symptomology (QIDS) derived from the IDS-SR in site two. Sleep disturbances were measured using three insomnia-related questions. Multilevel logistic regression was used to determine whether baseline insomnia scores were associated with TMS treatment outcome. Upon dichotomous categorization of the sample by insomnia and sleep-medication use, depression and sleep scores were analyzed across time using mixed repeated measures ANOVA. Results We found that sleep quality improves after TMS (p<.001) and correlates with improvement in non-insomnia related depression symptoms (r= .318, p<.001). We found that among those who had significant insomnia at baseline, those not using sleep medications had significantly worse post-treatment IDS-SR scores compared to those using sleep medications (p=. 021) despite no difference in final insomnia score. Discussion Together, our results suggest that while baseline insomnia is not associated with TMS effectiveness, treating insomnia may affect the trajectory of TMS therapy. Future prospective studies are needed to examine the effect of insomnia treatment alongside TMS for depression.
Collapse
Affiliation(s)
- Jamie Kweon
- Brain Stimulation Mechanisms Laboratory, Division of Depression and Anxiety Disorders, McLean Hospital, Belmont, MA, United States
| | - Andrew M. Fukuda
- Brain Stimulation Mechanisms Laboratory, Division of Depression and Anxiety Disorders, McLean Hospital, Belmont, MA, United States
- Department of Psychiatry, Harvard Medical School, Boston, MA, United States
| | - Asi P. Gobin
- Neuromodulation Research Facility, Butler Hospital, Providence, RI, United States
| | - Lamaan Haq
- Brain Stimulation Mechanisms Laboratory, Division of Depression and Anxiety Disorders, McLean Hospital, Belmont, MA, United States
| | - Linda L. Carpenter
- Neuromodulation Research Facility, Butler Hospital, Providence, RI, United States
- Department of Psychiatry and Human Behavior, Alpert Medical School of Brown University, Butler Hospital, Providence, RI, United States
| | - Joshua C. Brown
- Brain Stimulation Mechanisms Laboratory, Division of Depression and Anxiety Disorders, McLean Hospital, Belmont, MA, United States
- Department of Psychiatry, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
11
|
Ambron R. Synaptic sensitization in the anterior cingulate cortex sustains the consciousness of pain via synchronized oscillating electromagnetic waves. Front Hum Neurosci 2024; 18:1462211. [PMID: 39323956 PMCID: PMC11422113 DOI: 10.3389/fnhum.2024.1462211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Accepted: 08/29/2024] [Indexed: 09/27/2024] Open
Abstract
A recent report showed that experiencing pain requires not only activities in the brain, but also the generation of electric fields in a defined area of the anterior cingulate cortex (ACC). The present manuscript presents evidence that electromagnetic (EM) waves are also necessary. Action potentials (APs) encoding information about an injury stimulate thousands synapses on pyramidal neurons within the ACC resulting in the generation of synchronized oscillating (EM) waves and the activation of NMDA receptors. The latter induces a long-term potentiation (LTP) in the pyramidal dendrites that is necessary to experience both neuropathic and visceral pain. The LTP sensitizes transmission across the synapses that sustains the duration of the waves and the pain, EM waves containing information about the injury travel throughout the brain and studies using transcranial stimulation indicate that they can induce NMDA-mediated LTP in distant neuronal circuits. What is ultimately experienced as pain depends on the almost instantaneous integration of information from numerous neuronal centers, such as the amygdala, that are widely separated in the brain. These centers also generate EM waves and I propose that the EM waves from these centers interact to rapidly adjust the intensity of the pain to accommodate past and present circumstances. Where the waves are transformed into a consciousness of pain is unknown. One possibility is the mind which, according to contemporary theories, is where conscious experiences arise. The hypothesis can be tested directly by blocking the waves from the ACC. If correct, the waves would open new avenues of research into the relationship between the brain, consciousness, and the mind.
Collapse
|
12
|
Sundaram P, Dong C, Makaroff S, Okada Y. How conductivity boundaries influence the electric field induced by transcranial magnetic stimulation in in vitro experiments. Brain Stimul 2024; 17:1034-1044. [PMID: 39142380 PMCID: PMC11586064 DOI: 10.1016/j.brs.2024.08.003] [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: 05/03/2024] [Revised: 08/10/2024] [Accepted: 08/10/2024] [Indexed: 08/16/2024] Open
Abstract
BACKGROUND Although transcranial magnetic stimulation (TMS) has become a valuable method for non-invasive brain stimulation, the cellular basis of TMS activation of neurons is still not fully understood. In vitro preparations have been used to understand the biophysical mechanisms of TMS, but in many cases these studies have encountered substantial difficulties in activating neurons. OBJECTIVE/HYPOTHESIS The hypothesis of this work is that conductivity boundaries can have large effects on the electric field in commonly used in vitro preparations. Our goal was to analyze the resulting difficulties in in vitro TMS using a simulation study, using a charge-based boundary element model. METHODS We decomposed the total electric field into the sum of the primary electric field, which only depends on coil geometry and current, and the secondary electric field arising from conductivity boundaries, which strongly depends on tissue and chamber geometry. We investigated the effect of the conductivity boundaries on the electric field strength for a variety of in vitro experimental settings to determine the sources of difficulty. RESULTS We showed that conductivity boundaries can have large effects on the electric field in in vitro preparations. Depending on the geometry of the air-saline and the saline-tissue interfaces, the secondary electric field can significantly enhance, or attenuate the primary electric field, resulting in a much stronger or weaker total electric field inside the tissue; we showed this using a realistic preparation. Submerged chambers are generally much more efficient than interface chambers since the secondary field due to the thin film of saline covering the tissue in the interface chamber opposes the primary field and significantly reduces the total field in the tissue placed in the interface chamber. The relative dimensions of the chamber and the TMS coil critically determine the total field; the popular setup with a large coil and a small chamber is particularly sub-optimal because the secondary field due to the air-chamber boundary opposes the primary field, thereby attenuating the total field. The form factor (length vs width) of the tissue in the direction of the induced field can be important since a relatively narrow tissue enhances the total field at the saline-tissue boundary. CONCLUSIONS Overall, we found that the total electric field in the tissue is higher in submerged chambers, higher if the chamber size is larger than the coil and if the shorter tissue dimension is in the direction of the electric field. Decomposing the total field into the primary and secondary fields is useful for designing in vitro experiments and interpreting the results.
Collapse
Affiliation(s)
- Padmavathi Sundaram
- Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
| | - Chunling Dong
- Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Sergey Makaroff
- Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA; Worcester Polytechnic Institute, Worcester, MA, USA
| | - Yoshio Okada
- Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA; Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
13
|
Huang Y, Zelmann R, Hadar P, Dezha-Peralta J, Richardson RM, Williams ZM, Cash SS, Keller CJ, Paulk AC. Theta-burst direct electrical stimulation remodels human brain networks. Nat Commun 2024; 15:6982. [PMID: 39143083 PMCID: PMC11324911 DOI: 10.1038/s41467-024-51443-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Accepted: 08/07/2024] [Indexed: 08/16/2024] Open
Abstract
Theta-burst stimulation (TBS), a patterned brain stimulation technique that mimics rhythmic bursts of 3-8 Hz endogenous brain rhythms, has emerged as a promising therapeutic approach for treating a wide range of brain disorders, though the neural mechanism of TBS action remains poorly understood. We investigated the neural effects of TBS using intracranial EEG (iEEG) in 10 pre-surgical epilepsy participants undergoing intracranial monitoring. Here we show that individual bursts of direct electrical TBS at 29 frontal and temporal sites evoked strong neural responses spanning broad cortical regions. These responses exhibited dynamic local field potential voltage changes over the course of stimulation presentations, including either increasing or decreasing responses, suggestive of short-term plasticity. Stronger stimulation augmented the mean TBS response amplitude and spread with more recording sites demonstrating short-term plasticity. TBS responses were stimulation site-specific with stronger TBS responses observed in regions with strong baseline stimulation effective (cortico-cortical evoked potentials) and functional (low frequency phase locking) connectivity. Further, we could use these measures to predict stable and varying (e.g. short-term plasticity) TBS response locations. Future work may integrate pre-treatment connectivity alongside other biophysical factors to personalize stimulation parameters, thereby optimizing induction of neuroplasticity within disease-relevant brain networks.
Collapse
Affiliation(s)
- Yuhao Huang
- Department of Neurosurgery, Stanford University, Palo Alto, CA, USA
| | - Rina Zelmann
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Center for Neurotechnology and Neurorecovery, Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
| | - Peter Hadar
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Jaquelin Dezha-Peralta
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Center for Neurotechnology and Neurorecovery, Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
| | - R Mark Richardson
- Department of Neurosurgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Ziv M Williams
- Department of Neurosurgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Sydney S Cash
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Center for Neurotechnology and Neurorecovery, Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
| | - Corey J Keller
- Department of Psychiatry and Behavioral Sciences, Stanford University, Palo Alto, CA, USA.
- Wu Tsai Neurosciences Institute, Stanford University, Palo Alto, CA, USA.
- Veterans Affairs Palo Alto Healthcare System, and the Sierra Pacific Mental Illness, Research, Education, and Clinical Center (MIRECC), Palo Alto, CA, USA.
| | - Angelique C Paulk
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
- Center for Neurotechnology and Neurorecovery, Department of Neurology, Massachusetts General Hospital, Boston, MA, USA.
| |
Collapse
|
14
|
Lin J, Xing Q, Zhang C, Luo Y, Chen X, Xie Y, Wang Y. Advances in Repetitive Transcranial Magnetic Stimulation for the Treatment of Post-traumatic Stress Disorder. ALPHA PSYCHIATRY 2024; 25:440-448. [PMID: 39360295 PMCID: PMC11443297 DOI: 10.5152/alphapsychiatry.2024.241587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Accepted: 04/29/2024] [Indexed: 10/04/2024]
Abstract
Post-traumatic stress disorder (PTSD) is a psychiatric disorder that develops and persists after an individual experiences a major traumatic or life-threatening event. While pharmacological treatment and psychological interventions can alleviate some symptoms, pharmacotherapy is time-consuming with low patient compliance, and psychological interventions are costly. Repetitive Transcranial Magnetic Stimulation (rTMS) is a safe and effective technique for treating PTSD, with advantages such as high compliance, low cost, and simplicity of implementation. It can even simultaneously improve depressive symptoms in some patients. Current research indicates that high-frequency rTMS shows better therapeutic effects compared to low-frequency rTMS, with no significant difference in the likelihood of adverse reactions between the two. Theta Burst Stimulation (TBS) exhibits similar efficacy to high-frequency rTMS, with shorter duration and significant improvement in depressive symptoms. However, it carries a slightly higher risk of adverse reactions compared to traditional high-frequency rTMS. Combining rTMS with psychological therapy appears to be more effective in improving PTSD symptoms, with early onset of effects and longer duration, albeit at higher cost and requiring individualized patient control. The most common adverse effect of treatment is headache, which can be improved by stopping treatment or using analgesics. Despite these encouraging data, several aspects remain unknown. Given the highly heterogeneous nature of PTSD, defining unique treatment methods for this patient population is quite challenging. There are also considerable differences between trials regarding stimulation parameters, therapeutic effects, and the role of combined psychological therapy, which future research needs to address.
Collapse
Affiliation(s)
- Jingyi Lin
- Department of Rehabilitation Medicine, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
| | - Qijia Xing
- Department of Rehabilitation Medicine, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
| | - Chunyu Zhang
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Stem Cell Immunity and Regeneration Key Laboratory of Luzhou, Luzhou, Sichuan, China
| | - Yaomin Luo
- Department of Rehabilitation Medicine, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
| | - Xin Chen
- Department of Rehabilitation Medicine, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
| | - Yulei Xie
- Department of Rehabilitation Medicine, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
- School of Rehabilitation, Capital Medical University, Beijing, China
| | - Yinxu Wang
- Department of Rehabilitation Medicine, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
| |
Collapse
|
15
|
Tozzi L, Bertrand C, Hack LM, Lyons T, Olmsted AM, Rajasekharan D, Chen T, Berlow YA, Yesavage JA, Lim K, Madore MR, Philip NS, Holtzheimer P, Williams LM. A cognitive neural circuit biotype of depression showing functional and behavioral improvement after transcranial magnetic stimulation in the B-SMART-fMRI trial. NATURE. MENTAL HEALTH 2024; 2:987-998. [PMID: 39911692 PMCID: PMC11798407 DOI: 10.1038/s44220-024-00271-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Accepted: 05/16/2024] [Indexed: 02/07/2025]
Abstract
We previously identified a cognitive biotype of depression characterized by treatment resistance, impaired cognitive control behavioral performance and dysfunction in the cognitive control circuit, comprising the dorsolateral prefrontal cortex (dLPFC) and dorsal anterior cingulate cortex (dACC). Therapeutic transcranial magnetic stimulation (TMS) to the left dLPFC is a promising option for individuals whose depression does not respond to pharmacotherapy. Here, 43 veterans with treatment-resistant depression were assessed before TMS, after early TMS and post-TMS using functional magnetic resonance imaging during a Go-NoGo paradigm, behavioral cognitive control tests and symptom questionnaires. Stratifying veterans at baseline based on task-evoked dLPFC-dACC connectivity, we demonstrate that TMS-related improvement in cognitive control circuit connectivity and behavioral performance is specific to individuals with reduced connectivity at baseline (cognitive biotype +), whereas individuals with intact connectivity at baseline (cognitive biotype -) did not demonstrate significant changes. Our findings show that dLPFC-dACC connectivity during cognitive control is both a promising diagnostic biomarker for a cognitive biotype of depression and a response biomarker for cognitive improvement after TMS applied to the dLPFC.
Collapse
Affiliation(s)
- Leonardo Tozzi
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Claire Bertrand
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA, USA
- These authors contributed equally: Claire Bertrand, Laura Michele Hack, Timothy Lyons, Alisa Marie Olmsted, Divya Rajasekharan
| | - Laura Michele Hack
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA, USA
- Mental Illness Research, Education and Clinical Center, VA Palo Alto Health Care System, Palo Alto, CA, USA
- These authors contributed equally: Claire Bertrand, Laura Michele Hack, Timothy Lyons, Alisa Marie Olmsted, Divya Rajasekharan
| | - Timothy Lyons
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA, USA
- These authors contributed equally: Claire Bertrand, Laura Michele Hack, Timothy Lyons, Alisa Marie Olmsted, Divya Rajasekharan
| | - Alisa Marie Olmsted
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA, USA
- Mental Illness Research, Education and Clinical Center, VA Palo Alto Health Care System, Palo Alto, CA, USA
- These authors contributed equally: Claire Bertrand, Laura Michele Hack, Timothy Lyons, Alisa Marie Olmsted, Divya Rajasekharan
| | - Divya Rajasekharan
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA, USA
- These authors contributed equally: Claire Bertrand, Laura Michele Hack, Timothy Lyons, Alisa Marie Olmsted, Divya Rajasekharan
| | - TeChieh Chen
- National Center for PTSD, VA Medical Center, US Department of Veterans Affairs, White River Junction, VT, USA
| | - Yosef A. Berlow
- Department of Psychiatry and Human Behavior, Alpert Medical School of Brown University, Providence, RI, USA
- VA RR&D Center for Neurorestoration and Neurotechnology, VA Providence Healthcare System, Providence, RI, USA
| | - Jerome A. Yesavage
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA, USA
- Mental Illness Research, Education and Clinical Center, VA Palo Alto Health Care System, Palo Alto, CA, USA
| | - Kelvin Lim
- Department of Psychiatry and Behavioral Sciences, University of Minnesota Medical School, Minneapolis, MN, USA
- Minneapolis VA Health Care System, Minneapolis, MN, USA
- These authors jointly supervised this work: Kelvin Lim, Michelle Madore, Noah S. Philip, Paul Holtzheimer, Leanne Maree Williams
| | - Michelle R. Madore
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA, USA
- Mental Illness Research, Education and Clinical Center, VA Palo Alto Health Care System, Palo Alto, CA, USA
- These authors jointly supervised this work: Kelvin Lim, Michelle Madore, Noah S. Philip, Paul Holtzheimer, Leanne Maree Williams
| | - Noah S. Philip
- Department of Psychiatry and Human Behavior, Alpert Medical School of Brown University, Providence, RI, USA
- VA RR&D Center for Neurorestoration and Neurotechnology, VA Providence Healthcare System, Providence, RI, USA
- These authors jointly supervised this work: Kelvin Lim, Michelle Madore, Noah S. Philip, Paul Holtzheimer, Leanne Maree Williams
| | - Paul Holtzheimer
- National Center for PTSD, VA Medical Center, US Department of Veterans Affairs, White River Junction, VT, USA
- Geisel School of Medicine at Dartmouth, Hanover, NH, USA
- These authors jointly supervised this work: Kelvin Lim, Michelle Madore, Noah S. Philip, Paul Holtzheimer, Leanne Maree Williams
| | - Leanne Maree Williams
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA, USA
- Mental Illness Research, Education and Clinical Center, VA Palo Alto Health Care System, Palo Alto, CA, USA
- These authors jointly supervised this work: Kelvin Lim, Michelle Madore, Noah S. Philip, Paul Holtzheimer, Leanne Maree Williams
| |
Collapse
|
16
|
Kim H, Kornman PT, Kweon J, Wassermann EM, Wright DL, Li J, Brown JC. Combined effects of pharmacological interventions and intermittent theta-burst stimulation on motor sequence learning. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.24.604878. [PMID: 39211172 PMCID: PMC11361068 DOI: 10.1101/2024.07.24.604878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Drugs that modulate N-methyl-D-aspartate (NMDA) or γ-Aminobutyric acid type A (GABA A ) receptors can shed light on their role in synaptic plasticity mechanisms underlying the effects of non-invasive brain stimulation. However, research on the combined effects of these drugs and exogenous stimulation on motor learning is limited. This study aimed to investigate the effects of pharmacological interventions combined with intermittent theta-burst stimulation (iTBS) on human motor learning. Nine right-handed healthy subjects (mean age ± SD: 31.56 ± 12.96 years; 6 females) participated in this double-blind crossover study. All participants were assigned to four drug conditions in a randomized order: (1) D-cycloserine (partial NMDA receptor agonist), (2) D-cycloserine + dextromethorphan (NMDA receptor agonist + antagonist), (3) lorazepam (GABA A receptor agonist), and (4) placebo (identical microcrystalline cellulose capsule). After drug intake, participants practiced the 12-item keyboard sequential task as a baseline measure. Two hours after drug intake, iTBS was administered at the primary motor cortex. Following iTBS, the retention test was performed in the same manner as the baseline measure. Our findings revealed that lorazepam combined with iTBS impaired motor learning during the retention test. Future studies are still needed for a better understanding of the mechanisms through which TMS may influence human motor learning.
Collapse
|
17
|
Pecsok MK, Mordy A, Cristancho MA, Oathes D, Roalf DR. The Glutamatergic Effects of Clinical Repetitive Transcranial Magnetic Stimulation in Depressed Populations: A Preliminary Meta-Analysis of Proton Magnetic Resonance Spectroscopy Studies. Psychopathology 2024; 57:1-16. [PMID: 39004073 PMCID: PMC11724939 DOI: 10.1159/000538690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Accepted: 03/25/2024] [Indexed: 07/16/2024]
Abstract
INTRODUCTION Repetitive transcranial magnetic stimulation (rTMS) alleviates symptoms of major depressive disorder, but its neurobiological mechanisms remain to be fully understood. Growing evidence from proton magnetic resonance spectroscopy (1HMRS) studies suggests that rTMS alters excitatory and inhibitory neurometabolites. This preliminary meta-analysis aims to quantify current trends in the literature and identify future directions for the field. METHODS Ten eligible studies that quantified Glutamate (Glu), Glu+Glutamine (Glx), or GABA before and after an rTMS intervention in depressed samples were sourced from PubMed, MEDLINE, PsychInfo, Google Scholar, and primary literature following PRISMA guidelines. Data were pooled using a random-effects model, Cohen's d effect sizes were calculated, and moderators, such as neurometabolite and 1HMRS sequence, were assessed. It was hypothesized that rTMS would increase cortical neurometabolites. RESULTS Within-subjects data from 224 cases encompassing 31 neurometabolite effects (k) were analyzed. Active rTMS in clinical responders (n = 128; k = 22) nominally increased glutamatergic neurometabolites (d = 0.15 [95% CI: -0.01, 0.30], p = 0.06). No change was found in clinical nonresponders (p = 0.8) or sham rTMS participants (p = 0.4). A significant increase was identified in Glx (p = 0.01), but not Glu (p = 0.6). Importantly, effect size across conditions were associated with the number of rTMS pulses patients received (p = 0.05), suggesting dose dependence. CONCLUSIONS Clinical rTMS is associated with a nominal, dose-dependent increase in glutamatergic neurometabolites, suggesting rTMS may induce Glu-dependent neuroplasticity and upregulate neurometabolism. More, larger scale studies adhering to established acquisition and reporting standards are needed to further elucidate the neurometabolic mechanisms of rTMS.
Collapse
Affiliation(s)
- Maggie K Pecsok
- Department of Psychiatry, University of Pennsylvania, Philadelphia, Pennsylvania, USA,
| | - Arianna Mordy
- Department of Psychiatry, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Mario A Cristancho
- Department of Psychiatry, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Desmond Oathes
- Department of Psychiatry, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - David R Roalf
- Department of Psychiatry, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
18
|
Kweon J, Vigne M, Fukuda AM, Ren B, Carpenter LL, Brown JC. NMDA and GABA Receptor-Mediated Plasticity Induced by 10-Hz Repetitive Transcranial Magnetic Stimulation. RESEARCH SQUARE 2024:rs.3.rs-4630964. [PMID: 38978559 PMCID: PMC11230474 DOI: 10.21203/rs.3.rs-4630964/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
Although 10-Hz repetitive transcranial magnetic stimulation (rTMS) is an FDA-approved treatment for depression, we have yet to fully understand the mechanism through which rTMS induces therapeutic and durable changes in the brain. Two competing theories have emerged suggesting that 10-Hz rTMS induces N-methyl-D-aspartate receptor (NMDAR)-dependent long-term potentiation (LTP), or alternatively, removal of inhibitory gamma-aminobutyric acid receptors (GABARs). We examined these two proposed mechanisms of action in the human motor cortex in a double-blind, randomized, four-arm crossover study in healthy subjects. We tested motor-evoked potentials (MEPs) before and after 10-Hz rTMS in the presence of four drugs separated by 1-week each: placebo, NMDAR partial agonist d-cycloserine (DCS 100mg), DCS 100mg + NMDAR partial antagonist dextromethorphan (DMO 150mg; designed to "knock down" DCS-mediated facilitation), and GABAR agonist lorazepam (LZP 2.5mg). NMDAR agonism by DCS enhanced rTMS-induced cortical excitability more than placebo. This enhancement was blocked by combining DCS with NMDAR antagonist, DMO. If GABARs are removed by rTMS, GABAR agonism via LZP should lack its inhibitory effect yielding higher post/pre MEPs. However, MEPs were reduced after rTMS indicating stability of GABAR numbers. These data suggest that 10-Hz rTMS facilitation in the healthy motor cortex may enact change in the brain through NMDAR-mediated LTP-like mechanisms rather than through GABAergic reduction.
Collapse
Affiliation(s)
- Jamie Kweon
- Brain Stimulation Mechanisms Laboratory, Neurotherapeutics, Division of Depression and Anxiety, McLean Hospital
| | - Megan Vigne
- Department of Psychiatry and Human Behavior, Alpert Medical School of Brown University, Butler Hospital
| | - Andrew M Fukuda
- Brain Stimulation Mechanisms Laboratory, Neurotherapeutics, Division of Depression and Anxiety, McLean Hospital
| | - Boyu Ren
- Department of Psychiatry, Harvard Medical School
| | - Linda L Carpenter
- Department of Psychiatry and Human Behavior, Alpert Medical School of Brown University, Butler Hospital
| | - Joshua C Brown
- Brain Stimulation Mechanisms Laboratory, Neurotherapeutics, Division of Depression and Anxiety, McLean Hospital
| |
Collapse
|
19
|
Holl N, Heerdegen M, Zschorlich V, Köhling R, Kirschstein T. Inhibition of Acute mGluR5-Dependent Depression in Hippocampal CA1 by High-Frequency Magnetic Stimulation. Brain Sci 2024; 14:603. [PMID: 38928603 PMCID: PMC11202050 DOI: 10.3390/brainsci14060603] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2024] [Revised: 06/05/2024] [Accepted: 06/11/2024] [Indexed: 06/28/2024] Open
Abstract
High-frequency magnetic stimulation (HFMS) applied directly to the hippocampal slice preparation in vitro induces activity-dependent synaptic plasticity and metaplasticity. In addition, changes in synaptic transmission following HFMS involve the activation of N-methyl-D-aspartate and metabotropic glutamate receptors (mGluR). Here, we asked whether a short period of HFMS (5 × 10 delta-burst trains, duration of ~1 min) could alter mGluR5-mediated depression at Schaffer collateral-CA1 synapses in the acute brain slice preparation at 30 min after HFMS. To this end, we obtained field excitatory postsynaptic potential (fEPSP) slopes from Schaffer collateral-CA1 synapses after HFMS or control. First, we demonstrated that activity-dependent plasticity following HFMS depends on the slice orientation towards the magnetic coil indicating specific ion fluxes induced by magnetic fields. Second, we found that the mGluR5-specific agonist (RS)-2-chloro-5-hydroxyphenylglycine reduced the field excitatory postsynaptic potential (fEPSP) slopes in control slices but rather enhanced them in HFMS-treated slices. In contrast, the compound (S)-3,5-dihydroxyphenylglycine acting at both mGluR1 and mGluR5 reduced fEPSP slopes in both control and HFMS-treated slices. Importantly, the mGluR-dependent effects were independent from the slice-to-coil orientation indicating that asynchronous glutamate release could play a role. We conclude that a short period of HFMS inhibits subsequently evoked mGluR5-dependent depression at Schaffer collateral-CA1 synapses. This could be relevant for repetitive transcranial magnetic stimulation in psychiatric disorders such as major depression.
Collapse
Affiliation(s)
- Norman Holl
- Oscar Langendorff Institute of Physiology, University Medicine Rostock, University of Rostock, Gertrudenstrasse 9, 18057 Rostock, Germany; (N.H.); (M.H.); (R.K.)
| | - Marco Heerdegen
- Oscar Langendorff Institute of Physiology, University Medicine Rostock, University of Rostock, Gertrudenstrasse 9, 18057 Rostock, Germany; (N.H.); (M.H.); (R.K.)
| | - Volker Zschorlich
- Institute of Sport Sciences, University of Rostock, Am Waldessaum 23a, 18057 Rostock, Germany;
- Institute of Sport Sciences, Carl von Ossietzky University of Oldenburg, Ammerländer Heerstraße 114-118, 26129 Oldenburg, Germany
| | - Rüdiger Köhling
- Oscar Langendorff Institute of Physiology, University Medicine Rostock, University of Rostock, Gertrudenstrasse 9, 18057 Rostock, Germany; (N.H.); (M.H.); (R.K.)
| | - Timo Kirschstein
- Oscar Langendorff Institute of Physiology, University Medicine Rostock, University of Rostock, Gertrudenstrasse 9, 18057 Rostock, Germany; (N.H.); (M.H.); (R.K.)
| |
Collapse
|
20
|
Thai M, Nair AU, Klimes-Dougan B, Albott CS, Silamongkol T, Corkrum M, Hill D, Roemer JW, Lewis CP, Croarkin PE, Lim KO, Widge AS, Nahas Z, Eberly LE, Cullen KR. Deep transcranial magnetic stimulation for adolescents with treatment-resistant depression: A preliminary dose-finding study exploring safety and clinical effectiveness. J Affect Disord 2024; 354:589-600. [PMID: 38484878 PMCID: PMC11163675 DOI: 10.1016/j.jad.2024.03.061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 02/20/2024] [Accepted: 03/09/2024] [Indexed: 03/26/2024]
Abstract
BACKGROUND Transcranial magnetic stimulation (TMS) is an intervention for treatment-resistant depression (TRD) that modulates neural activity. Deep TMS (dTMS) can target not only cortical but also deeper limbic structures implicated in depression. Although TMS has demonstrated safety in adolescents, dTMS has yet to be applied to adolescent TRD. OBJECTIVE/HYPOTHESIS This pilot study evaluated the safety, tolerability, and clinical effects of dTMS in adolescents with TRD. We hypothesized dTMS would be safe, tolerable, and efficacious for adolescent TRD. METHODS 15 adolescents with TRD (Age, years: M = 16.4, SD = 1.42) completed a six-week daily dTMS protocol targeting the left dorsolateral prefrontal cortex (BrainsWay H1 coil, 30 sessions, 10 Hz, 3.6 s train duration, 20s inter-train interval, 55 trains; 1980 total pulses per session, 80 % to 120 % of motor threshold). Participants completed clinical, safety, and neurocognitive assessments before and after treatment. The primary outcome was depression symptom severity measured by the Children's Depression Rating Scale-Revised (CDRS-R). RESULTS 14 out of 15 participants completed the dTMS treatments. One participant experienced a convulsive syncope; the other participants only experienced mild side effects (e.g., headaches). There were no serious adverse events and minimal to no change in cognitive performance. Depression symptom severity significantly improved pre- to post-treatment and decreased to a clinically significant degree after 10 treatment sessions. Six participants met criteria for treatment response. LIMITATIONS Main limitations include a small sample size and open-label design. CONCLUSIONS These findings provide preliminary evidence that dTMS may be tolerable and associated with clinical improvement in adolescent TRD.
Collapse
Affiliation(s)
- Michelle Thai
- Department of Psychology, University of Minnesota, Twin Cities, MN, United States of America; Center for Depression, Anxiety, and Stress Research, McLean Hospital, Belmont, MA, United States of America; Department of Psychiatry, Harvard Medical School, United States of America.
| | - Aparna U Nair
- Department of Psychiatry & Behavioral Sciences, University of Minnesota Medical School, Minneapolis, MN, United States of America
| | - Bonnie Klimes-Dougan
- Department of Psychology, University of Minnesota, Twin Cities, MN, United States of America
| | - C Sophia Albott
- Department of Psychiatry & Behavioral Sciences, University of Minnesota Medical School, Minneapolis, MN, United States of America
| | - Thanharat Silamongkol
- Graduate School of Applied and Professional Psychology, Rutgers, The State University of New Jersey, New Brunswick, NJ, United States of America
| | - Michelle Corkrum
- Columbia University Medical Center, New York, NY, United States of America
| | - Dawson Hill
- University of Michigan Medical School, Ann Arbor, MI, United States of America
| | - Justin W Roemer
- Department of Psychiatry & Behavioral Sciences, University of Minnesota Medical School, Minneapolis, MN, United States of America
| | - Charles P Lewis
- Department of Psychiatry & Behavioral Sciences, University of Minnesota Medical School, Minneapolis, MN, United States of America
| | - Paul E Croarkin
- Department of Psychiatry and Psychology, Mayo Clinic, Rochester, MN, United States of America
| | - Kelvin O Lim
- Department of Psychiatry & Behavioral Sciences, University of Minnesota Medical School, Minneapolis, MN, United States of America
| | - Alik S Widge
- Department of Psychiatry & Behavioral Sciences, University of Minnesota Medical School, Minneapolis, MN, United States of America
| | - Ziad Nahas
- Department of Psychiatry & Behavioral Sciences, University of Minnesota Medical School, Minneapolis, MN, United States of America
| | - Lynn E Eberly
- Division of Biostatistics, School of Public Health, University of Minnesota, United States of America
| | - Kathryn R Cullen
- Department of Psychiatry & Behavioral Sciences, University of Minnesota Medical School, Minneapolis, MN, United States of America
| |
Collapse
|
21
|
Galanis C, Neuhaus L, Hananeia N, Turi Z, Jedlicka P, Vlachos A. Axon morphology and intrinsic cellular properties determine repetitive transcranial magnetic stimulation threshold for plasticity. Front Cell Neurosci 2024; 18:1374555. [PMID: 38638302 PMCID: PMC11025360 DOI: 10.3389/fncel.2024.1374555] [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: 01/22/2024] [Accepted: 03/13/2024] [Indexed: 04/20/2024] Open
Abstract
Introduction Repetitive transcranial magnetic stimulation (rTMS) is a widely used therapeutic tool in neurology and psychiatry, but its cellular and molecular mechanisms are not fully understood. Standardizing stimulus parameters, specifically electric field strength, is crucial in experimental and clinical settings. It enables meaningful comparisons across studies and facilitates the translation of findings into clinical practice. However, the impact of biophysical properties inherent to the stimulated neurons and networks on the outcome of rTMS protocols remains not well understood. Consequently, achieving standardization of biological effects across different brain regions and subjects poses a significant challenge. Methods This study compared the effects of 10 Hz repetitive magnetic stimulation (rMS) in entorhino-hippocampal tissue cultures from mice and rats, providing insights into the impact of the same stimulation protocol on similar neuronal networks under standardized conditions. Results We observed the previously described plastic changes in excitatory and inhibitory synaptic strength of CA1 pyramidal neurons in both mouse and rat tissue cultures, but a higher stimulation intensity was required for the induction of rMS-induced synaptic plasticity in rat tissue cultures. Through systematic comparison of neuronal structural and functional properties and computational modeling, we found that morphological parameters of CA1 pyramidal neurons alone are insufficient to explain the observed differences between the groups. Although morphologies of mouse and rat CA1 neurons showed no significant differences, simulations confirmed that axon morphologies significantly influence individual cell activation thresholds. Notably, differences in intrinsic cellular properties were sufficient to account for the 10% higher intensity required for the induction of synaptic plasticity in the rat tissue cultures. Conclusion These findings demonstrate the critical importance of axon morphology and intrinsic cellular properties in predicting the plasticity effects of rTMS, carrying valuable implications for the development of computer models aimed at predicting and standardizing the biological effects of rTMS.
Collapse
Affiliation(s)
- Christos Galanis
- Department of Neuroanatomy, Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Lena Neuhaus
- Department of Neuroanatomy, Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Nicholas Hananeia
- 3R-Zentrum Gießen, Justus-Liebig-Universitat Giessen, Giessen, Germany
| | - Zsolt Turi
- Department of Neuroanatomy, Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Peter Jedlicka
- 3R-Zentrum Gießen, Justus-Liebig-Universitat Giessen, Giessen, Germany
| | - Andreas Vlachos
- Department of Neuroanatomy, Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Center BrainLinks-BrainTools, University of Freiburg, Freiburg, Germany
- Center for Basics in NeuroModulation (NeuroModulBasics), Faculty of Medicine, University of Freiburg, Freiburg, Germany
| |
Collapse
|
22
|
Abstract
Repetitive transcranial magnetic stimulation (rTMS) has become an increasingly popular tool to modulate neural excitability and induce neural plasticity in clinical and preclinical models; however, the physiological mechanisms in which it exerts these effects remain largely unknown. To date, studies have primarily focused on characterizing rTMS-induced changes occurring at the synapse, with little attention given to changes in intrinsic membrane properties. However, accumulating evidence suggests that rTMS may induce its effects, in part, via intrinsic plasticity mechanisms, suggesting a new and potentially complementary understanding of how rTMS alters neural excitability and neural plasticity. In this review, we provide an overview of several intrinsic plasticity mechanisms before reviewing the evidence for rTMS-induced intrinsic plasticity. In addition, we discuss a select number of neurological conditions where rTMS-induced intrinsic plasticity has therapeutic potential before speculating on the temporal relationship between rTMS-induced intrinsic and synaptic plasticity.
Collapse
Affiliation(s)
- Emily S King
- Experimental and Regenerative Neurosciences, School of Biological Sciences, The University of Western Australia, Perth, Australia
- Perron Institute for Neurological and Translational Science, Perth, Australia
| | - Alexander D Tang
- Experimental and Regenerative Neurosciences, School of Biological Sciences, The University of Western Australia, Perth, Australia
- Perron Institute for Neurological and Translational Science, Perth, Australia
| |
Collapse
|
23
|
Liu L, Hu H, Wu J, Koleske AJ, Chen H, Wang N, Yu K, Wu Y, Xiao X, Zhang Q. Integrin α3 is required for high-frequency repetitive transcranial magnetic stimulation-induced glutamatergic synaptic transmission in mice with ischemia. CNS Neurosci Ther 2024; 30:e14498. [PMID: 37867481 PMCID: PMC11017422 DOI: 10.1111/cns.14498] [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: 06/05/2023] [Revised: 09/16/2023] [Accepted: 10/01/2023] [Indexed: 10/24/2023] Open
Abstract
BACKGROUND Repetitive transcranial magnetic stimulation (rTMS) is an effective therapy in post-stroke motor recovery. However, the underlying mechanisms of rTMS regulates long-lasting changes with synaptic transmission and glutamate receptors function (including AMPARs or NMDARs) remains unclear. METHODS Mice were received 10-Hz rTMS treatment once daily on the third day after photothrombotic (PT) stroke for 18 days. Motor behaviors and the Western blot were used to evaluate the therapeutic efficacy of 10-Hz rTMS in the mice with PT model. Moreover, we used wild-type (WT) and NEX-α3-/- mice to further explore the 10-Hz rTMS effect. RESULTS We found that 10-Hz rTMS improved the post-stroke motor performance in the PT mice. Moreover, the levels of AMPAR, vGlut1, and integrin α3 in the peri-infarct were significantly increased in the rTMS group. In contrast, 10-Hz rTMS did not induce these aforementioned effects in NEX-α3-/- mice. The amplitude of AMPAR-mediated miniature excitatory postsynaptic currents (EPSCs) and evoked EPSCs was increased in the WT + rTMS group, but did not change in NEX-α3-/- mice with rTMS. CONCLUSIONS In this study, 10-Hz rTMS improved the glutamatergic synaptic transmission in the peri-infract cortex through effects on integrin α3 and AMPARs, which resulted in motor function recovery after stroke.
Collapse
Affiliation(s)
- Li Liu
- Department of Rehabilitation Medicine, Huashan HospitalFudan UniversityShanghaiChina
| | - Han Hu
- Behavioral and Cognitive Neuroscience CenterInstitute of Science and Technology for Brain‐Inspired Intelligence, Fudan UniversityShanghaiChina
| | - Junfa Wu
- Department of Rehabilitation Medicine, Huashan HospitalFudan UniversityShanghaiChina
| | - Anthony J. Koleske
- Departments of Molecular Biophysics and Biochemistry and NeuroscienceYale UniversityNew HavenConnecticutUSA
| | - Hongting Chen
- Behavioral and Cognitive Neuroscience CenterInstitute of Science and Technology for Brain‐Inspired Intelligence, Fudan UniversityShanghaiChina
| | - Nianhong Wang
- Department of Rehabilitation Medicine, Huashan HospitalFudan UniversityShanghaiChina
| | - Kewei Yu
- Department of Rehabilitation Medicine, Huashan HospitalFudan UniversityShanghaiChina
| | - Yi Wu
- Department of Rehabilitation Medicine, Huashan HospitalFudan UniversityShanghaiChina
| | - Xiao Xiao
- Behavioral and Cognitive Neuroscience CenterInstitute of Science and Technology for Brain‐Inspired Intelligence, Fudan UniversityShanghaiChina
| | - Qun Zhang
- Department of Rehabilitation Medicine, Huashan HospitalFudan UniversityShanghaiChina
| |
Collapse
|
24
|
Ye H, Dima M, Hall V, Hendee J. Cellular mechanisms underlying carry-over effects after magnetic stimulation. Sci Rep 2024; 14:5167. [PMID: 38431662 PMCID: PMC10908793 DOI: 10.1038/s41598-024-55915-8] [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: 06/03/2023] [Accepted: 02/28/2024] [Indexed: 03/05/2024] Open
Abstract
Magnetic fields are widely used for neuromodulation in clinical settings. The intended effect of magnetic stimulation is that neural activity resumes its pre-stimulation state right after stimulation. Many theoretical and experimental works have focused on the cellular and molecular basis of the acute neural response to magnetic field. However, effects of magnetic stimulation can still last after the termination of the magnetic stimulation (named "carry-over effects"), which could generate profound effects to the outcome of the stimulation. However, the cellular and molecular mechanisms of carry-over effects are largely unknown, which renders the neural modulation practice using magnetic stimulation unpredictable. Here, we investigated carry-over effects at the cellular level, using the combination of micro-magnetic stimulation (µMS), electrophysiology, and computation modeling. We found that high frequency magnetic stimulation could lead to immediate neural inhibition in ganglion neurons from Aplysia californica, as well as persistent, carry-over inhibition after withdrawing the magnetic stimulus. Carry-over effects were found in the neurons that fired action potentials under a variety of conditions. The carry-over effects were also observed in the neurons when the magnetic field was applied across the ganglion sheath. The state of the neuron, specifically synaptic input and membrane potential fluctuation, plays a significant role in generating the carry-over effects after magnetic stimulation. To elucidate the cellular mechanisms of such carry-over effects under magnetic stimulation, we simulated a single neuron under magnetic stimulation with multi-compartment modeling. The model successfully replicated the carry-over effects in the neuron, and revealed that the carry-over effect was due to the dysfunction of the ion channel dynamics that were responsible for the initiation and sustaining of membrane excitability. A virtual voltage-clamp experiment revealed a compromised Na conductance and enhanced K conductance post magnetic stimulation, rendering the neurons incapable of generating action potentials and, therefore, leading to the carry over effects. Finally, both simulation and experimental results demonstrated that the carry-over effects could be controlled by disturbing the membrane potential during the post-stimulus inhibition period. Delineating the cellular and ion channel mechanisms underlying carry-over effects could provide insights to the clinical outcomes in brain stimulation using TMS and other modalities. This research incentivizes the development of novel neural engineering or pharmacological approaches to better control the carry-over effects for optimized clinical outcomes.
Collapse
Affiliation(s)
- Hui Ye
- Department of Biology, Loyola University Chicago, Quinlan Life Sciences Education and Research Center, 1032 W. Sheridan Rd., Chicago, IL, 60660, USA.
| | - Maria Dima
- Department of Biology, Loyola University Chicago, Quinlan Life Sciences Education and Research Center, 1032 W. Sheridan Rd., Chicago, IL, 60660, USA
| | - Vincent Hall
- Department of Biology, Loyola University Chicago, Quinlan Life Sciences Education and Research Center, 1032 W. Sheridan Rd., Chicago, IL, 60660, USA
| | - Jenna Hendee
- Department of Biology, Loyola University Chicago, Quinlan Life Sciences Education and Research Center, 1032 W. Sheridan Rd., Chicago, IL, 60660, USA
| |
Collapse
|
25
|
Zhu Y, Liao L, Gao S, Tao Y, Huang H, Fang X, Yuan C, Gao C. Neuroprotective effects of repetitive transcranial magnetic stimulation on Alzheimer's disease: Undetermined therapeutic protocols and mechanisms. NEUROPROTECTION 2024; 2:16-32. [DOI: 10.1002/nep3.40] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/11/2023] [Accepted: 01/24/2024] [Indexed: 01/03/2025]
Abstract
AbstractAlzheimer's disease (AD) is a prevalent neurodegenerative disorder characterized by gradual deterioration of cognitive functions, for which an effective treatment is currently unavailable. Repetitive transcranial magnetic stimulation (rTMS), a well‐established noninvasive brain stimulation method, is utilized in clinical settings to address various neuropsychiatric conditions, such as depression, neuropathic pain, and poststroke dysfunction. Increasing evidence suggests that rTMS may enhance cognitive abilities in individuals with AD. However, its optimal therapeutic protocols and precise mechanisms are currently unknown, impeding its clinical implementation. In the present review, we aimed to summarize and discuss the efficacy‐related parameters in rTMS treatment, encompassing stimulus frequency, stimulus pattern, stimulus intensity, and the configuration of the stimulus coil. Furthermore, we reviewed promising rTMS therapeutic protocols involving various combinations of these factors, that were examined in clinical studies. Based on our analysis, we propose that a multisite high‐frequency rTMS (HF‐rTMS) regimen has value in AD therapy, and that promising single‐site protocols, such as HF‐rTMS, applied over the left dorsolateral prefrontal cortex, precuneus, or cerebellum are required to be validated in larger clinical studies. Lastly, we provide a comprehensive review of the potential mechanisms underlying the neuroprotective effects of rTMS on cognition in AD in terms of brain network modulation as well as cellular and molecular reactions. In conclusion, the interaction of diverse mechanisms may be responsible for the total therapeutic effect of rTMS on AD. This review provides theoretical and practical evidence for the future clinical application and scientific research of rTMS in AD.
Collapse
Affiliation(s)
- Yang Zhu
- Department of Rehabilitation Medicine, Daping Hospital Army Medical University Chongqing China
| | - Lingyi Liao
- Department of Rehabilitation Medicine, Daping Hospital Army Medical University Chongqing China
| | - Shihao Gao
- Department of Rehabilitation Medicine, Daping Hospital Army Medical University Chongqing China
| | - Yong Tao
- Department of Rehabilitation Medicine, Daping Hospital Army Medical University Chongqing China
| | - Hao Huang
- Department of Rehabilitation Medicine, Daping Hospital Army Medical University Chongqing China
- Department of Rehabilitation Medicine General Hospital of Southern Theatre Command of PLA Guangzhou China
| | - Xiangqin Fang
- Department of Rehabilitation Medicine, Daping Hospital Army Medical University Chongqing China
| | - Changyan Yuan
- Department of Rehabilitation Medicine, Daping Hospital Army Medical University Chongqing China
| | - Changyue Gao
- Department of Rehabilitation Medicine, Daping Hospital Army Medical University Chongqing China
| |
Collapse
|
26
|
Kruse P, Brandes G, Hemeling H, Huang Z, Wrede C, Hegermann J, Vlachos A, Lenz M. Synaptopodin Regulates Denervation-Induced Plasticity at Hippocampal Mossy Fiber Synapses. Cells 2024; 13:114. [PMID: 38247806 PMCID: PMC10814840 DOI: 10.3390/cells13020114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 12/17/2023] [Accepted: 12/27/2023] [Indexed: 01/23/2024] Open
Abstract
Neurological diseases can lead to the denervation of brain regions caused by demyelination, traumatic injury or cell death. The molecular and structural mechanisms underlying lesion-induced reorganization of denervated brain regions, however, are a matter of ongoing investigation. In order to address this issue, we performed an entorhinal cortex lesion (ECL) in mouse organotypic entorhino-hippocampal tissue cultures of both sexes and studied denervation-induced plasticity of mossy fiber synapses, which connect dentate granule cells (dGCs) with CA3 pyramidal cells (CA3-PCs) and play important roles in learning and memory formation. Partial denervation caused a strengthening of excitatory neurotransmission in dGCs, CA3-PCs and their direct synaptic connections, as revealed by paired recordings (dGC-to-CA3-PC). These functional changes were accompanied by ultrastructural reorganization of mossy fiber synapses, which regularly contain the plasticity-regulating protein synaptopodin and the spine apparatus organelle. We demonstrate that the spine apparatus organelle and synaptopodin are related to ribosomes in close proximity to synaptic sites and reveal a synaptopodin-related transcriptome. Notably, synaptopodin-deficient tissue preparations that lack the spine apparatus organelle failed to express lesion-induced synaptic adjustments. Hence, synaptopodin and the spine apparatus organelle play a crucial role in regulating lesion-induced synaptic plasticity at hippocampal mossy fiber synapses.
Collapse
Affiliation(s)
- Pia Kruse
- Department of Neuroanatomy, Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany
| | - Gudrun Brandes
- Institute of Neuroanatomy and Cell Biology, Hannover Medical School, 30625 Hannover, Germany
| | - Hanna Hemeling
- Department of Neuroanatomy, Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany
| | - Zhong Huang
- Institute of Neuroanatomy and Cell Biology, Hannover Medical School, 30625 Hannover, Germany
| | - Christoph Wrede
- Institute of Functional and Applied Anatomy, Hannover Medical School, 30625 Hannover, Germany
- Research Core Unit Electron Microscopy, Hannover Medical School, 30625 Hannover, Germany
| | - Jan Hegermann
- Institute of Functional and Applied Anatomy, Hannover Medical School, 30625 Hannover, Germany
- Research Core Unit Electron Microscopy, Hannover Medical School, 30625 Hannover, Germany
| | - Andreas Vlachos
- Department of Neuroanatomy, Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany
- Center for Basics in Neuromodulation (NeuroModulBasics), Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
- Center BrainLinks-BrainTools, University of Freiburg, 79104 Freiburg, Germany
| | - Maximilian Lenz
- Department of Neuroanatomy, Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany
- Institute of Neuroanatomy and Cell Biology, Hannover Medical School, 30625 Hannover, Germany
| |
Collapse
|
27
|
Sohn MN, Brown JC, Sharma P, Ziemann U, McGirr A. Pharmacological adjuncts and transcranial magnetic stimulation-induced synaptic plasticity: a systematic review. J Psychiatry Neurosci 2024; 49:E59-E76. [PMID: 38359933 PMCID: PMC10890793 DOI: 10.1503/jpn.230090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 10/23/2023] [Accepted: 11/08/2023] [Indexed: 02/17/2024] Open
Abstract
BACKGROUND Transcranial magnetic stimulation (TMS) is a noninvasive neurostimulation modality that has been used to study human synaptic plasticity. Leveraging work in ex vivo preparations, mechanistically informed pharmacological adjuncts to TMS have been used to improve our fundamental understanding of TMS-induced synaptic plasticity. METHODS We systematically reviewed the literature pairing pharmacological adjuncts with TMS plasticity-induction protocols in humans. We searched MEDLINE, PsycINFO, and Embase from 2013 to Mar. 10, 2023. Studies published before 2013 were extracted from a previous systematic review. We included studies using repetitive TMS, theta-burst stimulation, paired associative stimulation, and quadripulse stimulation paradigms in healthy and clinical populations. RESULTS Thirty-six studies met our inclusion criteria (28 in healthy and 8 in clinical populations). Most pharmacological agents have targeted the glutamatergic N-methyl-d-aspartate (NMDA; 15 studies) or dopamine receptors (13 studies). The NMDA receptor is necessary for TMS-induced plasticity; however, sufficiency has not been shown across protocols. Dopaminergic modulation of TMS-induced plasticity appears to be dose-dependent. The GABAergic, cholinergic, noradrenergic, and serotonergic neurotransmitter systems have small evidence bases supporting modulation of TMS-induced plasticity, as do voltage-gated calcium and sodium channels. Studies in clinical populations suggest that pharmacological adjuncts to TMS may rescue motor cortex plasticity, with implications for therapeutic applications of TMS and a promising clinical trial in depression. LIMITATIONS This review is limited by the predominance in the literature of studies with small sample sizes and crossover designs. CONCLUSION Pharmacologically enhanced TMS largely parallels findings from ex vivo preparations. As this area expands and novel targets are tested, adequately powered samples in healthy and clinical populations will inform the mechanisms of TMS-induced plasticity in health and disease.
Collapse
Affiliation(s)
- Myren N Sohn
- From the Hotchkiss Brain Institute, University of Calgary, Calgary, Alta., Canada (Sohn, McGirr); the Department of Psychiatry, University of Calgary, Alta., Canada (Sohn, McGirr); the Mathison Centre for Mental Health Research and Education, Calgary, Alta., Canada (Sohn, McGirr); the McLean Hospital, Division of Neurotherapeutics, Belmont, Mass., USA (Brown, Sharma); the Department of Psychiatry, Harvard Medical School, Boston, Mass., USA (Brown); the Department of Neurology & Stroke, Eberhard-Karls University, Tübingen, Germany (Ziemann); and the Hertie-Institute for Clinical Brain Research, Eberhard-Karls University, Tübingen, Germany (Ziemann)
| | - Joshua C Brown
- From the Hotchkiss Brain Institute, University of Calgary, Calgary, Alta., Canada (Sohn, McGirr); the Department of Psychiatry, University of Calgary, Alta., Canada (Sohn, McGirr); the Mathison Centre for Mental Health Research and Education, Calgary, Alta., Canada (Sohn, McGirr); the McLean Hospital, Division of Neurotherapeutics, Belmont, Mass., USA (Brown, Sharma); the Department of Psychiatry, Harvard Medical School, Boston, Mass., USA (Brown); the Department of Neurology & Stroke, Eberhard-Karls University, Tübingen, Germany (Ziemann); and the Hertie-Institute for Clinical Brain Research, Eberhard-Karls University, Tübingen, Germany (Ziemann)
| | - Prayushi Sharma
- From the Hotchkiss Brain Institute, University of Calgary, Calgary, Alta., Canada (Sohn, McGirr); the Department of Psychiatry, University of Calgary, Alta., Canada (Sohn, McGirr); the Mathison Centre for Mental Health Research and Education, Calgary, Alta., Canada (Sohn, McGirr); the McLean Hospital, Division of Neurotherapeutics, Belmont, Mass., USA (Brown, Sharma); the Department of Psychiatry, Harvard Medical School, Boston, Mass., USA (Brown); the Department of Neurology & Stroke, Eberhard-Karls University, Tübingen, Germany (Ziemann); and the Hertie-Institute for Clinical Brain Research, Eberhard-Karls University, Tübingen, Germany (Ziemann)
| | - Ulf Ziemann
- From the Hotchkiss Brain Institute, University of Calgary, Calgary, Alta., Canada (Sohn, McGirr); the Department of Psychiatry, University of Calgary, Alta., Canada (Sohn, McGirr); the Mathison Centre for Mental Health Research and Education, Calgary, Alta., Canada (Sohn, McGirr); the McLean Hospital, Division of Neurotherapeutics, Belmont, Mass., USA (Brown, Sharma); the Department of Psychiatry, Harvard Medical School, Boston, Mass., USA (Brown); the Department of Neurology & Stroke, Eberhard-Karls University, Tübingen, Germany (Ziemann); and the Hertie-Institute for Clinical Brain Research, Eberhard-Karls University, Tübingen, Germany (Ziemann)
| | - Alexander McGirr
- From the Hotchkiss Brain Institute, University of Calgary, Calgary, Alta., Canada (Sohn, McGirr); the Department of Psychiatry, University of Calgary, Alta., Canada (Sohn, McGirr); the Mathison Centre for Mental Health Research and Education, Calgary, Alta., Canada (Sohn, McGirr); the McLean Hospital, Division of Neurotherapeutics, Belmont, Mass., USA (Brown, Sharma); the Department of Psychiatry, Harvard Medical School, Boston, Mass., USA (Brown); the Department of Neurology & Stroke, Eberhard-Karls University, Tübingen, Germany (Ziemann); and the Hertie-Institute for Clinical Brain Research, Eberhard-Karls University, Tübingen, Germany (Ziemann)
| |
Collapse
|
28
|
Shenoy S, Ibrahim S. Perinatal Depression and the Role of Synaptic Plasticity in Its Pathogenesis and Treatment. Behav Sci (Basel) 2023; 13:942. [PMID: 37998688 PMCID: PMC10669186 DOI: 10.3390/bs13110942] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Revised: 11/09/2023] [Accepted: 11/14/2023] [Indexed: 11/25/2023] Open
Abstract
Emerging evidence indicates that synaptic plasticity is significantly involved in the pathophysiology and treatment of perinatal depression. Animal models have demonstrated the effects of overstimulated or weakened synapses in various circuits of the brain in causing affective disturbances. GABAergic theory of depression, stress, and the neuroplasticity model of depression indicate the role of synaptic plasticity in the pathogenesis of depression. Multiple factors related to perinatal depression like hormonal shifts, newer antidepressants, mood stabilizers, monoamine systems, biomarkers, neurotrophins, cytokines, psychotherapy and electroconvulsive therapy have demonstrated direct and indirect effects on synaptic plasticity. In this review, we discuss and summarize the various patho-physiology-related effects of synaptic plasticity in depression. We also discuss the association of treatment-related aspects related to psychotropics, electroconvulsive therapy, neuromodulation, psychotherapy, physical exercise and yoga with synaptic plasticity in perinatal depression. Future insights into newer methods of treatment directed towards the modulation of neuroplasticity for perinatal depression will be discussed.
Collapse
Affiliation(s)
- Sonia Shenoy
- Department of Psychiatry, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India;
| | - Sufyan Ibrahim
- Neuro-Informatics Laboratory, Mayo Clinic, Rochester, MN 55902, USA
| |
Collapse
|
29
|
Dufor T, Lohof AM, Sherrard RM. Magnetic Stimulation as a Therapeutic Approach for Brain Modulation and Repair: Underlying Molecular and Cellular Mechanisms. Int J Mol Sci 2023; 24:16456. [PMID: 38003643 PMCID: PMC10671429 DOI: 10.3390/ijms242216456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 11/08/2023] [Accepted: 11/13/2023] [Indexed: 11/26/2023] Open
Abstract
Neurological and psychiatric diseases generally have no cure, so innovative non-pharmacological treatments, including non-invasive brain stimulation, are interesting therapeutic tools as they aim to trigger intrinsic neural repair mechanisms. A common brain stimulation technique involves the application of pulsed magnetic fields to affected brain regions. However, investigations of magnetic brain stimulation are complicated by the use of many different stimulation parameters. Magnetic brain stimulation is usually divided into two poorly connected approaches: (1) clinically used high-intensity stimulation (0.5-2 Tesla, T) and (2) experimental or epidemiologically studied low-intensity stimulation (μT-mT). Human tests of both approaches are reported to have beneficial outcomes, but the underlying biology is unclear, and thus optimal stimulation parameters remain ill defined. Here, we aim to bring together what is known about the biology of magnetic brain stimulation from human, animal, and in vitro studies. We identify the common effects of different stimulation protocols; show how different types of pulsed magnetic fields interact with nervous tissue; and describe cellular mechanisms underlying their effects-from intracellular signalling cascades, through synaptic plasticity and the modulation of network activity, to long-term structural changes in neural circuits. Recent advances in magneto-biology show clear mechanisms that may explain low-intensity stimulation effects in the brain. With its large breadth of stimulation parameters, not available to high-intensity stimulation, low-intensity focal magnetic stimulation becomes a potentially powerful treatment tool for human application.
Collapse
Affiliation(s)
- Tom Dufor
- Department of Cell and Developmental Biology, University College London, London WC1E 6BT, UK
| | - Ann M. Lohof
- Sorbonne Université and CNRS, UMR8256 Biological Adaptation and Ageing, 75005 Paris, France;
| | - Rachel M. Sherrard
- Sorbonne Université and CNRS, UMR8256 Biological Adaptation and Ageing, 75005 Paris, France;
| |
Collapse
|
30
|
Munot S, Kim N, Huang Y, Keller CJ. Direct cortical stimulation induces short-term plasticity of neural oscillations in humans. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.15.567302. [PMID: 38014071 PMCID: PMC10680685 DOI: 10.1101/2023.11.15.567302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2023]
Abstract
Patterned brain stimulation is commonly employed as a tool for eliciting plasticity in brain circuits and treating neuropsychiatric disorders. Although widely used in clinical settings, there remains a limited understanding of how stimulation-induced plasticity influences neural oscillations and their interplay with the underlying baseline functional architecture. To address this question, we applied 15 minutes of 10Hz focal electrical simulation, a pattern identical to 'excitatory' repetitive transcranial magnetic stimulation (rTMS), to 14 medically-intractable epilepsy patients undergoing intracranial electroencephalographic (iEEG). We quantified the spectral features of the cortico-cortical evoked potential (CCEPs) in these patients before and after stimulation. We hypothesized that for a given region the temporal and spectral components of the CCEP predicted the location and degree of stimulation-induced plasticity. Across patients, low frequency power (alpha and beta) showed the broadest change, while the magnitude of change was stronger in high frequencies (beta and gamma). Next we demonstrated that regions with stronger baseline evoked spectral responses were more likely to undergo plasticity after stimulation. These findings were specific to a given frequency in a specific temporal window. Post-stimulation power changes were driven by the interaction between direction of change in baseline power and temporal window of change. Finally, regions exhibiting early increases and late decreases in evoked baseline power exhibited power changes after stimulation and were independent of stimulation location. Together, these findings that time-frequency baseline features predict post-stimulation plasticity effects demonstrate properties akin to Hebbian learning in humans and extend this theory to the temporal and spectral window of interest. These findings can help improve our understanding of human brain plasticity and lead to more effective brain stimulation techniques.
Collapse
Affiliation(s)
- Saachi Munot
- Department of Psychiatry and Behavioral Sciences, Stanford University Medical Center, Stanford, CA, USA
- Wu Tsai Neuroscience Institute, Stanford University, Stanford, CA, USA
- Veterans Affairs Palo Alto Healthcare System, and the Sierra Pacific Mental Illness, Research, Education, and Clinical Center (MIRECC), Palo Alto, CA, USA
| | - Naryeong Kim
- Department of Psychiatry and Behavioral Sciences, Stanford University Medical Center, Stanford, CA, USA
- Wu Tsai Neuroscience Institute, Stanford University, Stanford, CA, USA
- Veterans Affairs Palo Alto Healthcare System, and the Sierra Pacific Mental Illness, Research, Education, and Clinical Center (MIRECC), Palo Alto, CA, USA
| | - Yuhao Huang
- Wu Tsai Neuroscience Institute, Stanford University, Stanford, CA, USA
- Department of Neurosurgery, Stanford University Medical Center, Stanford, CA, USA
| | - Corey J. Keller
- Department of Psychiatry and Behavioral Sciences, Stanford University Medical Center, Stanford, CA, USA
- Wu Tsai Neuroscience Institute, Stanford University, Stanford, CA, USA
- Veterans Affairs Palo Alto Healthcare System, and the Sierra Pacific Mental Illness, Research, Education, and Clinical Center (MIRECC), Palo Alto, CA, USA
| |
Collapse
|
31
|
Anil S, Lu H, Rotter S, Vlachos A. Repetitive transcranial magnetic stimulation (rTMS) triggers dose-dependent homeostatic rewiring in recurrent neuronal networks. PLoS Comput Biol 2023; 19:e1011027. [PMID: 37956202 PMCID: PMC10681319 DOI: 10.1371/journal.pcbi.1011027] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 11/27/2023] [Accepted: 10/11/2023] [Indexed: 11/15/2023] Open
Abstract
Repetitive transcranial magnetic stimulation (rTMS) is a non-invasive brain stimulation technique used to induce neuronal plasticity in healthy individuals and patients. Designing effective and reproducible rTMS protocols poses a major challenge in the field as the underlying biomechanisms of long-term effects remain elusive. Current clinical protocol designs are often based on studies reporting rTMS-induced long-term potentiation or depression of synaptic transmission. Herein, we employed computational modeling to explore the effects of rTMS on long-term structural plasticity and changes in network connectivity. We simulated a recurrent neuronal network with homeostatic structural plasticity among excitatory neurons, and demonstrated that this mechanism was sensitive to specific parameters of the stimulation protocol (i.e., frequency, intensity, and duration of stimulation). Particularly, the feedback-inhibition initiated by network stimulation influenced the net stimulation outcome and hindered the rTMS-induced structural reorganization, highlighting the role of inhibitory networks. These findings suggest a novel mechanism for the lasting effects of rTMS, i.e., rTMS-induced homeostatic structural plasticity, and highlight the importance of network inhibition in careful protocol design, standardization, and optimization of stimulation.
Collapse
Affiliation(s)
- Swathi Anil
- Department of Neuroanatomy, Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Bernstein Center Freiburg, University of Freiburg, Freiburg, Germany
- Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Han Lu
- Department of Neuroanatomy, Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Center BrainLinks-BrainTools, University of Freiburg, Freiburg, Germany
| | - Stefan Rotter
- Bernstein Center Freiburg, University of Freiburg, Freiburg, Germany
- Faculty of Biology, University of Freiburg, Freiburg, Germany
- Center BrainLinks-BrainTools, University of Freiburg, Freiburg, Germany
| | - Andreas Vlachos
- Department of Neuroanatomy, Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Bernstein Center Freiburg, University of Freiburg, Freiburg, Germany
- Center BrainLinks-BrainTools, University of Freiburg, Freiburg, Germany
- Center for Basics in NeuroModulation (NeuroModulBasics), Faculty of Medicine, University of Freiburg, Freiburg, Germany
| |
Collapse
|
32
|
Citrenbaum C, Corlier J, Ngo D, Vince-Cruz N, Wilson A, Wilke SA, Krantz D, Tadayonnejad R, Ginder N, Levitt J, Lee JH, Leuchter MK, Strouse TB, Corse A, Vyas P, Leuchter AF. Pretreatment pupillary reactivity is associated with differential early response to 10 Hz and intermittent theta-burst repetitive transcranial magnetic stimulation (rTMS) treatment of major depressive disorder (MDD). Brain Stimul 2023; 16:1566-1571. [PMID: 37863389 DOI: 10.1016/j.brs.2023.10.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 10/04/2023] [Accepted: 10/08/2023] [Indexed: 10/22/2023] Open
Abstract
BACKGROUND Repetitive Transcranial Magnetic Stimulation (rTMS) is an effective treatment for Major Depressive Disorder (MDD). Two common rTMS protocols, 10 Hz and intermittent theta burst stimulation (iTBS), have comparable rates of efficacy in groups of patients. Recent evidence suggests that some individuals may be more likely to benefit from one form of stimulation than the other. The pretreatment pupillary light reflex (PLR) is significantly associated with response to a full course of rTMS using heterogeneous stimulation protocols. OBJECTIVE To test whether the relationship between pretreatment PLR and early symptom improvement differed between subjects treated with iTBS or 10 Hz stimulation. METHODS PLR was measured in 52 subjects who received solely 10 Hz (n = 35) or iTBS (n = 17) to left dorsolateral prefrontal cortex (DLPFC) for the first ten sessions of their treatment course. Primary outcome measure was the percent change of Inventory of Depressive Symptomatology - Self Report (IDS-SR) from session 1 to session 10. RESULTS There was a positive association between normalized maximum constriction velocity (nMCV) and early improvement in subjects receiving 10 Hz stimulation (R = 0.48, p = 0.004) and a negative association in subjects receiving iTBS (R = -0.52, p = 0.03). ANOVA revealed a significant interaction between nMCV and the type of initial stimulation (p = 0.001). Among subjects with low nMCV, those initially treated with iTBS showed 2.6 times greater improvement after 10 sessions (p = 0.01) than subjects initially receiving 10 Hz stimulation. CONCLUSION nMCV may detect physiologic differences between those likely to benefit from 10 Hz or iTBS treatment. Future studies should examine whether PLR could guide prospective treatment selection.
Collapse
Affiliation(s)
- Cole Citrenbaum
- TMS Clinical and Research Program, Neuromodulation Division, Semel Institute for Neuroscience and Human Behavior at UCLA, Los Angeles, CA, USA; Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90024, USA
| | - Juliana Corlier
- TMS Clinical and Research Program, Neuromodulation Division, Semel Institute for Neuroscience and Human Behavior at UCLA, Los Angeles, CA, USA; Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90024, USA
| | - Doan Ngo
- TMS Clinical and Research Program, Neuromodulation Division, Semel Institute for Neuroscience and Human Behavior at UCLA, Los Angeles, CA, USA; Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90024, USA
| | - Nikita Vince-Cruz
- TMS Clinical and Research Program, Neuromodulation Division, Semel Institute for Neuroscience and Human Behavior at UCLA, Los Angeles, CA, USA; Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90024, USA
| | - Andrew Wilson
- Cooperative Institute for Research in Environmental Sciences (CIRES), University of Colorado Boulder, Boulder, CO, USA; NOAA National Centers for Environmental Information (NCEI), Boulder, CO, USA
| | - Scott A Wilke
- TMS Clinical and Research Program, Neuromodulation Division, Semel Institute for Neuroscience and Human Behavior at UCLA, Los Angeles, CA, USA; Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90024, USA
| | - David Krantz
- TMS Clinical and Research Program, Neuromodulation Division, Semel Institute for Neuroscience and Human Behavior at UCLA, Los Angeles, CA, USA; Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90024, USA
| | - Reza Tadayonnejad
- TMS Clinical and Research Program, Neuromodulation Division, Semel Institute for Neuroscience and Human Behavior at UCLA, Los Angeles, CA, USA; Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90024, USA; Division of the Humanities and Social Sciences, California Institute of Technology, Pasadena, CA, USA
| | - Nathaniel Ginder
- TMS Clinical and Research Program, Neuromodulation Division, Semel Institute for Neuroscience and Human Behavior at UCLA, Los Angeles, CA, USA; Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90024, USA
| | - Jennifer Levitt
- TMS Clinical and Research Program, Neuromodulation Division, Semel Institute for Neuroscience and Human Behavior at UCLA, Los Angeles, CA, USA; Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90024, USA
| | - John H Lee
- TMS Clinical and Research Program, Neuromodulation Division, Semel Institute for Neuroscience and Human Behavior at UCLA, Los Angeles, CA, USA; Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90024, USA
| | - Michael K Leuchter
- TMS Clinical and Research Program, Neuromodulation Division, Semel Institute for Neuroscience and Human Behavior at UCLA, Los Angeles, CA, USA; Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90024, USA
| | - Thomas B Strouse
- TMS Clinical and Research Program, Neuromodulation Division, Semel Institute for Neuroscience and Human Behavior at UCLA, Los Angeles, CA, USA; Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90024, USA
| | - Andrew Corse
- TMS Clinical and Research Program, Neuromodulation Division, Semel Institute for Neuroscience and Human Behavior at UCLA, Los Angeles, CA, USA; Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90024, USA
| | - Pooja Vyas
- Department of Psychiatry, University of California San Diego, San Diego, CA, USA
| | - Andrew F Leuchter
- TMS Clinical and Research Program, Neuromodulation Division, Semel Institute for Neuroscience and Human Behavior at UCLA, Los Angeles, CA, USA; Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90024, USA.
| |
Collapse
|
33
|
Galanis C, Neuhaus L, Hananeia N, Turi Z, Jedlicka P, Vlachos A. Axon morphology and intrinsic cellular properties determine repetitive transcranial magnetic stimulation threshold for plasticity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.25.559399. [PMID: 37808716 PMCID: PMC10557586 DOI: 10.1101/2023.09.25.559399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/10/2023]
Abstract
Repetitive transcranial magnetic stimulation (rTMS) is a widely used therapeutic tool in neurology and psychiatry, but its cellular and molecular mechanisms are not fully understood. Standardizing stimulus parameters, specifically electric field strength and direction, is crucial in experimental and clinical settings. It enables meaningful comparisons across studies and facilitating the translation of findings into clinical practice. However, the impact of biophysical properties inherent to the stimulated neurons and networks on the outcome of rTMS protocols remains not well understood. Consequently, achieving standardization of biological effects across different brain regions and subjects poses a significant challenge. This study compared the effects of 10 Hz repetitive magnetic stimulation (rMS) in entorhino-hippocampal tissue cultures from mice and rats, providing insights into the impact of the same stimulation protocol on similar neuronal networks under standardized conditions. We observed the previously described plastic changes in excitatory and inhibitory synaptic strength of CA1 pyramidal neurons in both mouse and rat tissue cultures, but a higher stimulation intensity was required for the induction of rMS-induced synaptic plasticity in rat tissue cultures. Through systematic comparison of neuronal structural and functional properties and computational modeling, we found that morphological parameters of CA1 pyramidal neurons alone are insufficient to explain the observed differences between the groups. However, axon morphologies of individual cells played a significant role in determining activation thresholds. Notably, differences in intrinsic cellular properties were sufficient to account for the 10 % higher intensity required for the induction of synaptic plasticity in the rat tissue cultures. These findings demonstrate the critical importance of axon morphology and intrinsic cellular properties in predicting the plasticity effects of rTMS, carrying valuable implications for the development of computer models aimed at predicting and standardizing the biological effects of rTMS.
Collapse
|
34
|
Meneses-San Juan D, Lamas M, Ramírez-Rodríguez GB. Repetitive Transcranial Magnetic Stimulation Reduces Depressive-like Behaviors, Modifies Dendritic Plasticity, and Generates Global Epigenetic Changes in the Frontal Cortex and Hippocampus in a Rodent Model of Chronic Stress. Cells 2023; 12:2062. [PMID: 37626872 PMCID: PMC10453847 DOI: 10.3390/cells12162062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 07/19/2023] [Accepted: 07/24/2023] [Indexed: 08/27/2023] Open
Abstract
Depression is the most common affective disorder worldwide, accounting for 4.4% of the global population, a figure that could increase in the coming decades. In depression, there exists a reduction in the availability of dendritic spines in the frontal cortex (FC) and hippocampus (Hp). In addition, histone modification and DNA methylation are also dysregulated epigenetic mechanisms in depression. Repetitive transcranial magnetic stimulation (rTMS) is a technique that is used to treat depression. However, the epigenetic mechanisms of its therapeutic effect are still not known. Therefore, in this study, we evaluated the antidepressant effect of 5 Hz rTMS and examined its effect on dendritic remodeling, immunoreactivity of synapse proteins, histone modification, and DNA methylation in the FC and Hp in a model of chronic mild stress. Our data indicated that stress generated depressive-like behaviors and that rTMS reverses this effect, romotes the formation of dendritic spines, and favors the presynaptic connection in the FC and DG (dentate gyrus), in addition to increasing histone H3 trimethylation and DNA methylation. These results suggest that the antidepressant effect of rTMS is associated with dendritic remodeling, which is probably regulated by epigenetic mechanisms. These data are a first approximation of the impact of rTMS at the epigenetic level in the context of depression. Therefore, it is necessary to analyze in future studies as to which genes are regulated by these mechanisms, and how they are associated with the neuroplastic modifications promoted by rTMS.
Collapse
Affiliation(s)
- David Meneses-San Juan
- National Institute of Psychiatry “Ramón de la Fuente Muñiz”, Mexico City 14370, Mexico;
- Center of Research and Advanced Studies of the National Polytechnic Institute, Mexico City 07360, Mexico;
| | - Mónica Lamas
- Center of Research and Advanced Studies of the National Polytechnic Institute, Mexico City 07360, Mexico;
| | | |
Collapse
|
35
|
Lenz M, Eichler A, Kruse P, Galanis C, Kleidonas D, Andrieux G, Boerries M, Jedlicka P, Müller U, Deller T, Vlachos A. The Amyloid Precursor Protein Regulates Synaptic Transmission at Medial Perforant Path Synapses. J Neurosci 2023; 43:5290-5304. [PMID: 37369586 PMCID: PMC10359033 DOI: 10.1523/jneurosci.1824-22.2023] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2022] [Revised: 05/20/2023] [Accepted: 05/24/2023] [Indexed: 06/29/2023] Open
Abstract
The perforant path provides the primary cortical excitatory input to the hippocampus. Because of its important role in information processing and coding, entorhinal projections to the dentate gyrus have been studied in considerable detail. Nevertheless, synaptic transmission between individual connected pairs of entorhinal stellate cells and dentate granule cells remains to be characterized. Here, we have used mouse organotypic entorhino-hippocampal tissue cultures of either sex, in which the entorhinal cortex (EC) to dentate granule cell (GC; EC-GC) projection is present, and EC-GC pairs can be studied using whole-cell patch-clamp recordings. By using cultures of wild-type mice, the properties of EC-GC synapses formed by afferents from the lateral and medial entorhinal cortex were compared, and differences in short-term plasticity were identified. As the perforant path is severely affected in Alzheimer's disease, we used tissue cultures of amyloid precursor protein (APP)-deficient mice to examine the role of APP at this synapse. APP deficiency altered excitatory neurotransmission at medial perforant path synapses, which was accompanied by transcriptomic and ultrastructural changes. Moreover, presynaptic but not postsynaptic APP deletion through the local injection of Cre-expressing adeno-associated viruses in conditional APPflox/flox tissue cultures increased the neurotransmission efficacy at perforant path synapses. In summary, these data suggest a physiological role for presynaptic APP at medial perforant path synapses that may be adversely affected under altered APP processing conditions.SIGNIFICANCE STATEMENT The hippocampus receives input from the entorhinal cortex via the perforant path. These projections to hippocampal dentate granule cells are of utmost importance for learning and memory formation. Although there is detailed knowledge about perforant path projections, the functional synaptic properties at the level of individual connected pairs of neurons are not well understood. In this study, we investigated the role of APP in mediating functional properties and transmission rules in individually connected neurons using paired whole-cell patch-clamp recordings and genetic tools in organotypic tissue cultures. Our results show that presynaptic APP expression limits excitatory neurotransmission via the perforant path, which could be compromised in pathologic conditions such as Alzheimer's disease.
Collapse
Affiliation(s)
- Maximilian Lenz
- Department of Neuroanatomy, Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany
- Hannover Medical School, Institute of Neuroanatomy and Cell Biology, 30625 Hannover, Germany
| | - Amelie Eichler
- Department of Neuroanatomy, Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany
| | - Pia Kruse
- Department of Neuroanatomy, Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany
| | - Christos Galanis
- Department of Neuroanatomy, Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany
| | - Dimitrios Kleidonas
- Department of Neuroanatomy, Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany
- Spemann Graduate School of Biology and Medicine, University of Freiburg, 79104 Freiburg, Germany
- Faculty of Biology, University of Freiburg, 79104 Freiburg, Germany
| | - Geoffroy Andrieux
- Institute of Medical Bioinformatics and Systems Medicine, Medical Center, Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany
| | - Melanie Boerries
- Institute of Medical Bioinformatics and Systems Medicine, Medical Center, Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany
- German Cancer Consortium, German Cancer Research Center, 69120 Heidelberg, Germany
| | - Peter Jedlicka
- Interdisciplinary Centre for 3Rs in Animal Research, Faculty of Medicine, Justus-Liebig-University, 35392 Giessen, Germany
- Institute of Clinical Neuroanatomy, Neuroscience Center, Goethe University Frankfurt, 60590 Frankfurt am Main, Germany
- Frankfurt Institute for Advanced Studies, 60438 Frankfurt am Main, Germany
| | - Ulrike Müller
- Institute of Pharmacy and Molecular Biotechnology, Functional Genomics, Ruprecht-Karls University Heidelberg, 69120 Heidelberg, Germany
| | - Thomas Deller
- Institute of Clinical Neuroanatomy, Neuroscience Center, Goethe University Frankfurt, 60590 Frankfurt am Main, Germany
| | - Andreas Vlachos
- Department of Neuroanatomy, Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany
- Center for Basics in Neuromodulation, Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany
- Center BrainLinks-BrainTools, University of Freiburg, 79104 Freiburg, Germany
| |
Collapse
|
36
|
Zhou L, Jin Y, Wu D, Cun Y, Zhang C, Peng Y, Chen N, Yang X, Zhang S, Ning R, Kuang P, Wang Z, Zhang P. Current evidence, clinical applications, and future directions of transcranial magnetic stimulation as a treatment for ischemic stroke. Front Neurosci 2023; 17:1177283. [PMID: 37534033 PMCID: PMC10390744 DOI: 10.3389/fnins.2023.1177283] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Accepted: 06/28/2023] [Indexed: 08/04/2023] Open
Abstract
Transcranial magnetic stimulation (TMS) is a non-invasive brain neurostimulation technique that can be used as one of the adjunctive treatment techniques for neurological recovery after stroke. Animal studies have shown that TMS treatment of rats with middle cerebral artery occlusion (MCAO) model reduced cerebral infarct volume and improved neurological dysfunction in model rats. In addition, clinical case reports have also shown that TMS treatment has positive neuroprotective effects in stroke patients, improving a variety of post-stroke neurological deficits such as motor function, swallowing, cognitive function, speech function, central post-stroke pain, spasticity, and other post-stroke sequelae. However, even though numerous studies have shown a neuroprotective effect of TMS in stroke patients, its possible neuroprotective mechanism is not clear. Therefore, in this review, we describe the potential mechanisms of TMS to improve neurological function in terms of neurogenesis, angiogenesis, anti-inflammation, antioxidant, and anti-apoptosis, and provide insight into the current clinical application of TMS in multiple neurological dysfunctions in stroke. Finally, some of the current challenges faced by TMS are summarized and some suggestions for its future research directions are made.
Collapse
Affiliation(s)
- Li Zhou
- Key Laboratory of Acupuncture and Massage for Treatment of Encephalopathy, College of Acupuncture, Tuina and Rehabilitation, Yunnan University of Traditional Chinese Medicine, Kunming, China
| | - Yaju Jin
- Key Laboratory of Acupuncture and Massage for Treatment of Encephalopathy, College of Acupuncture, Tuina and Rehabilitation, Yunnan University of Traditional Chinese Medicine, Kunming, China
| | - Danli Wu
- Key Laboratory of Acupuncture and Massage for Treatment of Encephalopathy, College of Acupuncture, Tuina and Rehabilitation, Yunnan University of Traditional Chinese Medicine, Kunming, China
| | - Yongdan Cun
- Key Laboratory of Acupuncture and Massage for Treatment of Encephalopathy, College of Acupuncture, Tuina and Rehabilitation, Yunnan University of Traditional Chinese Medicine, Kunming, China
| | - Chengcai Zhang
- Key Laboratory of Acupuncture and Massage for Treatment of Encephalopathy, College of Acupuncture, Tuina and Rehabilitation, Yunnan University of Traditional Chinese Medicine, Kunming, China
| | - Yicheng Peng
- Key Laboratory of Acupuncture and Massage for Treatment of Encephalopathy, College of Acupuncture, Tuina and Rehabilitation, Yunnan University of Traditional Chinese Medicine, Kunming, China
| | - Na Chen
- Key Laboratory of Acupuncture and Massage for Treatment of Encephalopathy, College of Acupuncture, Tuina and Rehabilitation, Yunnan University of Traditional Chinese Medicine, Kunming, China
| | - Xichen Yang
- Key Laboratory of Acupuncture and Massage for Treatment of Encephalopathy, College of Acupuncture, Tuina and Rehabilitation, Yunnan University of Traditional Chinese Medicine, Kunming, China
| | - Simei Zhang
- Key Laboratory of Acupuncture and Massage for Treatment of Encephalopathy, College of Acupuncture, Tuina and Rehabilitation, Yunnan University of Traditional Chinese Medicine, Kunming, China
| | - Rong Ning
- Key Laboratory of Acupuncture and Massage for Treatment of Encephalopathy, College of Acupuncture, Tuina and Rehabilitation, Yunnan University of Traditional Chinese Medicine, Kunming, China
| | - Peng Kuang
- Key Laboratory of Acupuncture and Massage for Treatment of Encephalopathy, College of Acupuncture, Tuina and Rehabilitation, Yunnan University of Traditional Chinese Medicine, Kunming, China
| | - Zuhong Wang
- Kunming Municipal Hospital of Traditional Chinese Medicine, Kunming, Yunnan, China
| | - Pengyue Zhang
- Key Laboratory of Acupuncture and Massage for Treatment of Encephalopathy, College of Acupuncture, Tuina and Rehabilitation, Yunnan University of Traditional Chinese Medicine, Kunming, China
| |
Collapse
|
37
|
Cano M, Lee E, Polanco C, Barbour T, Ellard KK, Andreou B, Uribe S, Henry ME, Seiner S, Cardoner N, Soriano-Mas C, Camprodon JA. Brain volumetric correlates of electroconvulsive therapy versus transcranial magnetic stimulation for treatment-resistant depression. J Affect Disord 2023; 333:140-146. [PMID: 37024015 PMCID: PMC10288116 DOI: 10.1016/j.jad.2023.03.093] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 03/19/2023] [Accepted: 03/29/2023] [Indexed: 04/08/2023]
Abstract
BACKGROUND Electroconvulsive therapy (ECT) and repetitive transcranial magnetic stimulation (rTMS) are effective neuromodulation therapies for treatment-resistant depression (TRD). While ECT is generally considered the most effective antidepressant, rTMS is less invasive, better tolerated and leads to more durable therapeutic benefits. Both interventions are established device antidepressants, but it remains unknown if they share a common mechanism of action. Here we aimed to compare the brain volumetric changes in patients with TRD after right unilateral (RUL) ECT versus left dorsolateral prefrontal cortex (lDLPFC) rTMS. METHODS We assessed 32 patients with TRD before the first treatment session and after treatment completion using structural magnetic resonance imaging. Fifteen patients were treated with RUL ECT and seventeen patients received lDLPFC rTMS. RESULTS Patients receiving RUL ECT, in comparison with patients treated with lDLPFC rTMS, showed a greater volumetric increase in the right striatum, pallidum, medial temporal lobe, anterior insular cortex, anterior midbrain, and subgenual anterior cingulate cortex. However, ECT- or rTMS-induced brain volumetric changes were not associated with the clinical improvement. LIMITATIONS We evaluated a modest sample size with concurrent pharmacological treatment and without neuromodulation therapies randomization. CONCLUSIONS Our findings suggest that despite comparable clinical outcomes, only RUL ECT is associated with structural change, while rTMS is not. We hypothesize that structural neuroplasticity and/or neuroinflammation may explain the larger structural changes observed after ECT, whereas neurophysiological plasticity may underlie the rTMS effects. More broadly, our results support the notion that there are multiple therapeutic strategies to move patients from depression to euthymia.
Collapse
Affiliation(s)
- Marta Cano
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA; Sant Pau Mental Health Research Group, Institut d'Investigació Biomèdica Sant Pau (IIB-Sant Pau), Hospital de la Santa Creu i Sant Pau, Barcelona, Spain; CIBERSAM, Carlos III Health Institute, Madrid, Spain
| | - Erik Lee
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Christopher Polanco
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Tracy Barbour
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Kristen K Ellard
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Blake Andreou
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Sofia Uribe
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Michael E Henry
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Stephen Seiner
- McLean Hospital, Harvard Medical School, Belmont, MA, USA
| | - Narcís Cardoner
- Sant Pau Mental Health Research Group, Institut d'Investigació Biomèdica Sant Pau (IIB-Sant Pau), Hospital de la Santa Creu i Sant Pau, Barcelona, Spain; CIBERSAM, Carlos III Health Institute, Madrid, Spain; Department of Psychiatry and Forensic Medicine, School of Medicine Bellaterra, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Carles Soriano-Mas
- CIBERSAM, Carlos III Health Institute, Madrid, Spain; Department of Psychiatry, Bellvitge University Hospital-IDIBELL, L'Hospitalet de Llobregat, Barcelona, Spain; Department of Social Psychology and Quantitative Psychology, University of Barcelona, Barcelona, Spain.
| | - Joan A Camprodon
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
38
|
Lokai T, Albin B, Qubbaj K, Tiwari AP, Adhikari P, Yang IH. A review on current brain organoid technologies from a biomedical engineering perspective. Exp Neurol 2023; 367:114461. [PMID: 37295544 DOI: 10.1016/j.expneurol.2023.114461] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 05/24/2023] [Accepted: 06/02/2023] [Indexed: 06/12/2023]
Abstract
Brain organoids are 3D cytoarchitectures resembling the embryonic human brain. This review focuses on current advancements in biomedical engineering methods to develop organoids such as pluripotent stem cells assemblies, quickly aggregated floating culture, hydrogel suspension, microfluidic systems (both photolithography and 3D printing), and brain organoids-on-a-chip. These methods have the potential to create a large impact on neurological disorder studies by creating a model of the human brain investigating pathogenesis and drug screening for individual patients. 3D brain organoid cultures mimic not only features of patients' unknown drug reactions, but also early human brain development at cellular, structural, and functional levels. The challenge of current brain organoids lies in the formation of distinct cortical neuron layers, gyrification, and the establishment of complex neuronal circuitry, as they are critically specialized, developmental aspects. Furthermore, recent advances such as vascularization and genome engineering are in development to overcome the barrier of neuronal complexity. Future technology of brain organoids is needed to improve tissue cross-communication, body axis simulation, cell patterning signals, and spatial-temporal control of differentiation, as engineering methods discussed in this review are rapidly evolving.
Collapse
Affiliation(s)
- Taylor Lokai
- Center for Biomedical Engineering and Science, Mechanical Engineering and Engineering Science, The University of North Carolina at Charlotte, Charlotte, NC 28223, USA
| | - Bayne Albin
- Center for Biomedical Engineering and Science, Mechanical Engineering and Engineering Science, The University of North Carolina at Charlotte, Charlotte, NC 28223, USA
| | - Khayzaran Qubbaj
- Center for Biomedical Engineering and Science, Mechanical Engineering and Engineering Science, The University of North Carolina at Charlotte, Charlotte, NC 28223, USA
| | - Arjun Prasad Tiwari
- Center for Biomedical Engineering and Science, Mechanical Engineering and Engineering Science, The University of North Carolina at Charlotte, Charlotte, NC 28223, USA
| | - Prashant Adhikari
- Center for Biomedical Engineering and Science, Mechanical Engineering and Engineering Science, The University of North Carolina at Charlotte, Charlotte, NC 28223, USA
| | - In Hong Yang
- Center for Biomedical Engineering and Science, Mechanical Engineering and Engineering Science, The University of North Carolina at Charlotte, Charlotte, NC 28223, USA.
| |
Collapse
|
39
|
Zeljkovic Jovanovic M, Stanojevic J, Stevanovic I, Stekic A, Bolland SJ, Jasnic N, Ninkovic M, Zaric Kontic M, Ilic TV, Rodger J, Nedeljkovic N, Dragic M. Intermittent Theta Burst Stimulation Improves Motor and Behavioral Dysfunction through Modulation of NMDA Receptor Subunit Composition in Experimental Model of Parkinson's Disease. Cells 2023; 12:1525. [PMID: 37296646 PMCID: PMC10252812 DOI: 10.3390/cells12111525] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 04/24/2023] [Accepted: 05/09/2023] [Indexed: 06/12/2023] Open
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disorder characterized by the progressive degeneration of the dopaminergic system, leading to a variety of motor and nonmotor symptoms. The currently available symptomatic therapy loses efficacy over time, indicating the need for new therapeutic approaches. Repetitive transcranial magnetic stimulation (rTMS) has emerged as one of the potential candidates for PD therapy. Intermittent theta burst stimulation (iTBS), an excitatory protocol of rTMS, has been shown to be beneficial in several animal models of neurodegeneration, including PD. The aim of this study was to investigate the effects of prolonged iTBS on motor performance and behavior and the possible association with changes in the NMDAR subunit composition in the 6-hydroxydopamine (6-OHDA)-induced experimental model of PD. Two-month-old male Wistar rats were divided into four groups: controls, 6-OHDA rats, 6-OHDA + iTBS protocol (two times/day/three weeks) and the sham group. The therapeutic effect of iTBS was evaluated by examining motor coordination, balance, spontaneous forelimb use, exploratory behavior, anxiety-like, depressive/anhedonic-like behavior and short-term memory, histopathological changes and changes at the molecular level. We demonstrated the positive effects of iTBS at both motor and behavioral levels. In addition, the beneficial effects were reflected in reduced degeneration of dopaminergic neurons and a subsequent increase in the level of DA in the caudoputamen. Finally, iTBS altered protein expression and NMDAR subunit composition, suggesting a sustained effect. Applied early in the disease course, the iTBS protocol may be a promising candidate for early-stage PD therapy, affecting motor and nonmotor deficits.
Collapse
Affiliation(s)
- Milica Zeljkovic Jovanovic
- Laboratory for Neurobiology, Department for General Physiology and Biophysics, Faculty of Biology, University of Belgrade, 11000 Belgrade, Serbia
| | - Jelena Stanojevic
- Institute for Medical Research, Military Medical Academy, 11000 Belgrade, Serbia
| | - Ivana Stevanovic
- Institute for Medical Research, Military Medical Academy, 11000 Belgrade, Serbia
- Medical Faculty of Military Medical Academy, University of Defense, 11000 Belgrade, Serbia
| | - Andjela Stekic
- Laboratory for Neurobiology, Department for General Physiology and Biophysics, Faculty of Biology, University of Belgrade, 11000 Belgrade, Serbia
| | - Samuel J. Bolland
- School of Biological Sciences, The University of Western Australia, Perth, WA 6009, Australia
- Perron Institute for Neurological and Translational Science, Perth, WA 6009, Australia
| | - Nebojsa Jasnic
- Department for Comparative Physiology and Ecophysiology, Faculty of Biology, University of Belgrade, 11000 Belgrade, Serbia
| | - Milica Ninkovic
- Institute for Medical Research, Military Medical Academy, 11000 Belgrade, Serbia
- Medical Faculty of Military Medical Academy, University of Defense, 11000 Belgrade, Serbia
| | - Marina Zaric Kontic
- Department of Molecular Biology and Endocrinology, Vinca Institute of Nuclear Sciences-National Institute of the Republic of Serbia, University of Belgrade, 11000 Belgrade, Serbia
| | - Tihomir V. Ilic
- Medical Faculty of Military Medical Academy, University of Defense, 11000 Belgrade, Serbia
| | - Jennifer Rodger
- School of Biological Sciences, The University of Western Australia, Perth, WA 6009, Australia
- Perron Institute for Neurological and Translational Science, Perth, WA 6009, Australia
| | - Nadezda Nedeljkovic
- Laboratory for Neurobiology, Department for General Physiology and Biophysics, Faculty of Biology, University of Belgrade, 11000 Belgrade, Serbia
| | - Milorad Dragic
- Laboratory for Neurobiology, Department for General Physiology and Biophysics, Faculty of Biology, University of Belgrade, 11000 Belgrade, Serbia
| |
Collapse
|
40
|
Eichler A, Kleidonas D, Turi Z, Fliegauf M, Kirsch M, Pfeifer D, Masuda T, Prinz M, Lenz M, Vlachos A. Microglial Cytokines Mediate Plasticity Induced by 10 Hz Repetitive Magnetic Stimulation. J Neurosci 2023; 43:3042-3060. [PMID: 36977586 PMCID: PMC10146500 DOI: 10.1523/jneurosci.2226-22.2023] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Revised: 02/15/2023] [Accepted: 03/10/2023] [Indexed: 03/29/2023] Open
Abstract
Microglia, the resident immune cells of the CNS, sense the activity of neurons and regulate physiological brain functions. They have been implicated in the pathology of brain diseases associated with alterations in neural excitability and plasticity. However, experimental and therapeutic approaches that modulate microglia function in a brain region-specific manner have not been established. In this study, we tested for the effects of repetitive transcranial magnetic stimulation (rTMS), a clinically used noninvasive brain stimulation technique, on microglia-mediated synaptic plasticity; 10 Hz electromagnetic stimulation triggered a release of plasticity-promoting cytokines from microglia in mouse organotypic brain tissue cultures of both sexes, while no significant changes in microglial morphology or microglia dynamics were observed. Indeed, substitution of tumor necrosis factor α (TNFα) and interleukin 6 (IL6) preserved synaptic plasticity induced by 10 Hz stimulation in the absence of microglia. Consistent with these findings, in vivo depletion of microglia abolished rTMS-induced changes in neurotransmission in the mPFC of anesthetized mice of both sexes. We conclude that rTMS affects neural excitability and plasticity by modulating the release of cytokines from microglia.SIGNIFICANCE STATEMENT Repetitive transcranial magnetic stimulation (rTMS) is a noninvasive brain stimulation technique that induces cortical plasticity. Despite its wide use in neuroscience and clinical practice (e.g., depression treatment), the cellular and molecular mechanisms of rTMS-mediated plasticity remain not well understood. Herein, we report an important role of microglia and plasticity-promoting cytokines in synaptic plasticity induced by 10 Hz rTMS in organotypic slice cultures and anesthetized mice, thereby identifying microglia-mediated synaptic adaptation as a target of rTMS-based interventions.
Collapse
Affiliation(s)
- Amelie Eichler
- Department of Neuroanatomy, Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany
| | - Dimitrios Kleidonas
- Department of Neuroanatomy, Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany
- Spemann Graduate School of Biology and Medicine, University of Freiburg, 79104 Freiburg, Germany
- Faculty of Biology, University of Freiburg, 79104 Freiburg, Germany
| | - Zsolt Turi
- Department of Neuroanatomy, Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany
| | - Maximilian Fliegauf
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
- Department of Pharmaceutical Biology and Biotechnology, Institute of Pharmaceutical Sciences, University of Freiburg, 79104 Freiburg, Germany
| | - Matthias Kirsch
- Department of Neuroanatomy, Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany
| | - Dietmar Pfeifer
- Department of Hematology, Oncology and Stem Cell Transplantation, Medical Center, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Takahiro Masuda
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
- Department of Molecular and System Pharmacology, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, 812-8582, Japan
| | - Marco Prinz
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, 79104 Freiburg, Germany
- Center for Basics in Neuromodulation (NeuroModulBasics), Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Maximilian Lenz
- Department of Neuroanatomy, Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany
| | - Andreas Vlachos
- Department of Neuroanatomy, Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany
- Center for Basics in Neuromodulation (NeuroModulBasics), Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
- Center BrainLinks-BrainTools, University of Freiburg, 79110 Freiburg, Germany
| |
Collapse
|
41
|
Kahnt T. Computationally Informed Interventions for Targeting Compulsive Behaviors. Biol Psychiatry 2023; 93:729-738. [PMID: 36464521 PMCID: PMC9989040 DOI: 10.1016/j.biopsych.2022.08.028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 08/04/2022] [Accepted: 08/30/2022] [Indexed: 11/02/2022]
Abstract
Compulsive behaviors are central to addiction and obsessive-compulsive disorder and can be understood as a failure of adaptive decision making. Particularly, they can be conceptualized as an imbalance in behavioral control, such that behavior is guided predominantly by learned rather than inferred outcome expectations. Inference is a computational process required for adaptive behavior, and recent work across species has identified the neural circuitry that supports inference-based decision making. This includes the orbitofrontal cortex, which has long been implicated in disorders of compulsive behavior. Inspired by evidence that modulating orbitofrontal cortex activity can alter inference-based behaviors, here we discuss noninvasive approaches to target these circuits in humans. Specifically, we discuss the potential of network-targeted transcranial magnetic stimulation and real-time neurofeedback to modulate the neural underpinnings of inference. Both interventions leverage recent advances in our understanding of the neurocomputational mechanisms of inference-based behavior and may be used to complement current treatment approaches for behavioral disorders.
Collapse
Affiliation(s)
- Thorsten Kahnt
- National Institute on Drug Abuse Intramural Research Program, Baltimore, Maryland.
| |
Collapse
|
42
|
Lin SHN, Lien YR, Shibata K, Sasaki Y, Watanabe T, Lin CP, Chang LH. The phase of plasticity-induced neurochemical changes of high-frequency repetitive transcranial magnetic stimulation are different from visual perceptual learning. Sci Rep 2023; 13:5720. [PMID: 37029245 PMCID: PMC10082079 DOI: 10.1038/s41598-023-32985-8] [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: 12/06/2022] [Accepted: 04/05/2023] [Indexed: 04/09/2023] Open
Abstract
Numerous studies have found that repetitive transcranial magnetic stimulation (rTMS) modulates plasticity. rTMS has often been used to change neural networks underlying learning, often under the assumption that the mechanism of rTMS-induced plasticity should be highly similar to that associated with learning. The presence of visual perceptual learning (VPL) reveals the plasticity of early visual systems, which is formed through multiple phases. Hence, we tested how high-frequency (HF) rTMS and VPL modulate the effect of visual plasticity by investigating neurometabolic changes in early visual areas. We employed an excitatory-to-inhibitory (E/I) ratio, which refers to glutamate concentration divided by GABA+ concentration, as an index of the degree of plasticity. We compared neurotransmitter concentration changes after applying HF rTMS to the visual cortex with those after training in a visual task, in otherwise identical procedures. Both the time courses of the E/I ratios and neurotransmitter contributions to the E/I ratio significantly differed between HF rTMS and training conditions. The peak E/I ratio occurred 3.5 h after HF rTMS with decreased GABA+, whereas the peak E/I ratio occurred 0.5 h after visual training with increased glutamate. Furthermore, HF rTMS temporally decreased the thresholds for detecting phosphene and perceiving low-contrast stimuli, indicating increased visual plasticity. These results suggest that plasticity in early visual areas induced by HF rTMS is not as involved in the early phase of development of VPL that occurs during and immediately after training.
Collapse
Affiliation(s)
- Shang-Hua N Lin
- Institute of Neuroscience, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Yun R Lien
- Institute of Neuroscience, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | | | - Yuka Sasaki
- Department of Cognitive, Linguistics, and Psychological Sciences, Brown University, Providence, USA
| | - Takeo Watanabe
- Department of Cognitive, Linguistics, and Psychological Sciences, Brown University, Providence, USA
| | - Ching-Po Lin
- Institute of Neuroscience, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Li-Hung Chang
- Institute of Neuroscience, National Yang Ming Chiao Tung University, Taipei, Taiwan.
- Institute of Philosophy of Mind and Cognition, National Yang Ming Chiao Tung University, Taipei, Taiwan.
| |
Collapse
|
43
|
Jiang W, Isenhart R, Liu CY, Song D. A C-shaped miniaturized coil for transcranial magnetic stimulation in rodents. J Neural Eng 2023; 20:026022. [PMID: 36863013 PMCID: PMC10037933 DOI: 10.1088/1741-2552/acc097] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 02/09/2023] [Accepted: 03/02/2023] [Indexed: 03/04/2023]
Abstract
Objective.Transcranial magnetic stimulation (TMS) is a non-invasive technique widely used for neuromodulation. Animal models are essential for investigating the underlying mechanisms of TMS. However, the lack of miniaturized coils hinders the TMS studies in small animals, since most commercial coils are designed for humans and thus incapable of focal stimulation in small animals. Furthermore, it is difficult to perform electrophysiological recordings at the TMS focal point using conventional coils.Approach.We designed, fabricated, and tested a novel miniaturized TMS coil (4-by-7 mm) that consisted of a C-shaped iron powder core and insulated copper wires (30 turns). The resulting magnetic and electric fields were characterized with experimental measurements and finite element modeling. The efficacy of this coil in neuromodulation was validated with electrophysiological recordings of single-unit activities (SUAs), somatosensory evoked potentials (SSEPs), and motor evoked potentials (MEPs) in rats (n= 32) following repetitive TMS (rTMS; 3 min, 10 Hz).Main results.This coil could generate a maximum magnetic field of 460 mT and an electric field of 7.2 V m-1in the rat brain according to our simulations. With subthreshold rTMS focally delivered over the sensorimotor cortex, mean firing rates of primary somatosensory and motor cortical neurons significantly increased (154±5% and 160±9% from the baseline level, respectively); MEP and SSEP amplitude significantly increased (136±9%) and decreased (74±4%), respectively.Significance.This miniaturized C-shaped coil enabled focal TMS and concurrent electrophysiological recording/stimulation at the TMS focal point. It provided a useful tool to investigate the neural responses and underlying mechanisms of TMS in small animal models. Using this paradigm, we for the first time observed distinct modulatory effects on SUAs, SSEPs, and MEPs with the same rTMS protocol in anesthetized rats. These results suggested that multiple neurobiological mechanisms in the sensorimotor pathways were differentially modulated by rTMS.
Collapse
Affiliation(s)
- Wenxuan Jiang
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA, United States of America
| | - Robert Isenhart
- Rancho Los Amigos National Rehabilitation Center, Downey, CA, United States of America
- USC Neurorestoration Center, University of Southern California, Los Angeles, CA, United States of America
| | - Charles Y Liu
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA, United States of America
- Rancho Los Amigos National Rehabilitation Center, Downey, CA, United States of America
- USC Neurorestoration Center, University of Southern California, Los Angeles, CA, United States of America
- Department of Neurological Surgery, University of Southern California, Los Angeles, CA, United States of America
| | - Dong Song
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA, United States of America
- USC Neurorestoration Center, University of Southern California, Los Angeles, CA, United States of America
| |
Collapse
|
44
|
Anil S, Lu H, Rotter S, Vlachos A. Repetitive transcranial magnetic stimulation (rTMS) triggers dose-dependent homeostatic rewiring in recurrent neuronal networks. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.20.533396. [PMID: 36993387 PMCID: PMC10055183 DOI: 10.1101/2023.03.20.533396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/31/2023]
Abstract
Repetitive transcranial magnetic stimulation (rTMS) is a non-invasive brain stimulation technique used to induce neuronal plasticity in healthy individuals and patients. Designing effective and reproducible rTMS protocols poses a major challenge in the field as the underlying biomechanisms remain elusive. Current clinical protocol designs are often based on studies reporting rTMS-induced long-term potentiation or depression of synaptic transmission. Herein, we employed computational modeling to explore the effects of rTMS on long-term structural plasticity and changes in network connectivity. We simulated a recurrent neuronal network with homeostatic structural plasticity between excitatory neurons, and demonstrated that this mechanism was sensitive to specific parameters of the stimulation protocol (i.e., frequency, intensity, and duration of stimulation). The feedback-inhibition initiated by network stimulation influenced the net stimulation outcome and hindered the rTMS-induced homeostatic structural plasticity, highlighting the role of inhibitory networks. These findings suggest a novel mechanism for the lasting effects of rTMS, i.e., rTMS-induced homeostatic structural plasticity, and highlight the importance of network inhibition in careful protocol design, standardization, and optimization of stimulation.
Collapse
Affiliation(s)
- Swathi Anil
- Department of Neuroanatomy, Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Bernstein Center Freiburg, University of Freiburg, Freiburg, Germany
- Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Han Lu
- Department of Neuroanatomy, Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Center BrainLinks-BrainTools, University of Freiburg, Freiburg, Germany
| | - Stefan Rotter
- Bernstein Center Freiburg, University of Freiburg, Freiburg, Germany
- Center BrainLinks-BrainTools, University of Freiburg, Freiburg, Germany
- Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Andreas Vlachos
- Department of Neuroanatomy, Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Bernstein Center Freiburg, University of Freiburg, Freiburg, Germany
- Center BrainLinks-BrainTools, University of Freiburg, Freiburg, Germany
- Center for Basics in NeuroModulation (NeuroModulBasics), Faculty of Medicine, University of Freiburg, Freiburg, Germany
| |
Collapse
|
45
|
Sherif MA, Khalil MZ, Shukla R, Brown JC, Carpenter LL. Synapses, predictions, and prediction errors: A neocortical computational study of MDD using the temporal memory algorithm of HTM. Front Psychiatry 2023; 14:976921. [PMID: 36911109 PMCID: PMC9995817 DOI: 10.3389/fpsyt.2023.976921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Accepted: 01/16/2023] [Indexed: 02/25/2023] Open
Abstract
Introduction Synapses and spines play a significant role in major depressive disorder (MDD) pathophysiology, recently highlighted by the rapid antidepressant effect of ketamine and psilocybin. According to the Bayesian brain and interoception perspectives, MDD is formalized as being stuck in affective states constantly predicting negative energy balance. To understand how spines and synapses relate to the predictive function of the neocortex and thus to symptoms, we used the temporal memory (TM), an unsupervised machine-learning algorithm. TM models a single neocortical layer, learns in real-time, and extracts and predicts temporal sequences. TM exhibits neocortical biological features such as sparse firing and continuous online learning using local Hebbian-learning rules. Methods We trained a TM model on random sequences of upper-case alphabetical letters, representing sequences of affective states. To model depression, we progressively destroyed synapses in the TM model and examined how that affected the predictive capacity of the network. We found that the number of predictions decreased non-linearly. Results Destroying 50% of the synapses slightly reduced the number of predictions, followed by a marked drop with further destruction. However, reducing the synapses by 25% distinctly dropped the confidence in the predictions. Therefore, even though the network was making accurate predictions, the network was no longer confident about these predictions. Discussion These findings explain how interoceptive cortices could be stuck in limited affective states with high prediction error. Connecting ketamine and psilocybin's proposed mechanism of action to depression pathophysiology, the growth of new synapses would allow representing more futuristic predictions with higher confidence. To our knowledge, this is the first study to use the TM model to connect changes happening at synaptic levels to the Bayesian formulation of psychiatric symptomatology. Linking neurobiological abnormalities to symptoms will allow us to understand the mechanisms of treatments and possibly, develop new ones.
Collapse
Affiliation(s)
- Mohamed A. Sherif
- Lifespan Physician Group, Department of Psychiatry and Human Behavior, The Warren Alpert Medical School of Brown University, Carney Institute for Brain Science, Norman Prince Neurosciences Institute, Providence, RI, United States
| | - Mostafa Z. Khalil
- Department of Psychiatry and Behavioral Health, Penn State Milton S. Hershey Medical Center, Penn State College of Medicine, Hershey, PA, United States
| | - Rammohan Shukla
- Department of Neurosciences, The University of Toledo College of Medicine and Life Sciences, Toledo, OH, United States
| | - Joshua C. Brown
- Department of Psychiatry and Human Behavior, The Warren Alpert Medical School of Brown University, Butler Hospital, Providence, RI, United States
| | - Linda L. Carpenter
- Department of Psychiatry and Human Behavior, The Warren Alpert Medical School of Brown University, Butler Hospital, Providence, RI, United States
| |
Collapse
|
46
|
Vigne M, Kweon J, Sharma P, Greenberg BD, Carpenter LL, Brown JC. Chronic caffeine consumption curbs rTMS-induced plasticity. Front Psychiatry 2023; 14:1137681. [PMID: 36911138 PMCID: PMC9993245 DOI: 10.3389/fpsyt.2023.1137681] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Accepted: 02/07/2023] [Indexed: 02/24/2023] Open
Abstract
Background Caffeine is a widely used psychostimulant. In the brain, caffeine acts as a competitive, non-selective adenosine receptor antagonist of A1 and A2A, both known to modulate long-term potentiation (LTP), the cellular basis of learning and memory. Repetitive transcranial magnetic stimulation (rTMS) is theorized to work through LTP induction and can modulate cortical excitability as measured by motor evoked potentials (MEPs). The acute effects of single caffeine doses diminish rTMS-induced corticomotor plasticity. However, plasticity in chronic daily caffeine users has not been examined. Method We conducted a post hoc secondary covariate analysis from two previously published plasticity-inducing pharmaco-rTMS studies combining 10 Hz rTMS and D-cycloserine (DCS) in twenty healthy subjects. Results In this hypothesis-generating pilot study, we observed enhanced MEP facilitation in non-caffeine users compared to caffeine users and placebo. Conclusion These preliminary data highlight a need to directly test the effects of caffeine in prospective well-powered studies, because in theory, they suggest that chronic caffeine use could limit learning or plasticity, including rTMS effectiveness.
Collapse
Affiliation(s)
- Megan Vigne
- Neuromodulation Research Facility, TMS Clinic, Butler Hospital, Providence, RI, United States
| | - Jamie Kweon
- Neuromodulation Research Facility, TMS Clinic, Butler Hospital, Providence, RI, United States
| | - Prayushi Sharma
- Neuromodulation Research Facility, TMS Clinic, Butler Hospital, Providence, RI, United States
| | - Benjamin D. Greenberg
- Neuromodulation Research Facility, TMS Clinic, Butler Hospital, Providence, RI, United States
- Department of Psychiatry and Human Behavior, Warren Alpert Medical School of Brown University, Providence, RI, United States
| | - Linda L. Carpenter
- Neuromodulation Research Facility, TMS Clinic, Butler Hospital, Providence, RI, United States
- Department of Psychiatry and Human Behavior, Warren Alpert Medical School of Brown University, Providence, RI, United States
| | - Joshua C. Brown
- Neuromodulation Research Facility, TMS Clinic, Butler Hospital, Providence, RI, United States
- Department of Psychiatry and Human Behavior, Warren Alpert Medical School of Brown University, Providence, RI, United States
- Department of Neurology, Warren Alpert Medical School of Brown University, Providence, RI, United States
| |
Collapse
|
47
|
Chen N, Qiu X, Hua Y, Hu J, Bai Y. Effects of sequential inhibitory and facilitatory repetitive transcranial magnetic stimulation on neurological and functional recovery of a patient with chronic stroke: A case report and literature review. Front Neurol 2023; 14:1064718. [PMID: 36779047 PMCID: PMC9911674 DOI: 10.3389/fneur.2023.1064718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Accepted: 01/05/2023] [Indexed: 01/28/2023] Open
Abstract
Background and purpose The effects of conventional protocols of repetitive transcranial magnetic stimulation (rTMS) in the chronic phase of stroke are limited. This study aimed to apply the sequential inhibitory and facilitatory rTMS for upper limb motor dysfunction post-stroke to observe the efficacy and explore the possible neurophysiological mechanism. We hypothesize that this protocol would both enhance the excitability of affected M1 and promote connections among motor areas. Case description We reported a 55-year-old female patient with a 1-year chronic stroke and right-sided hemiplegia, who underwent the 14-session rTMS with seven sessions of low frequency (LF) and with seven sessions of high frequency (HF). Clinical scales mainly including Fugl-Meyer Assessment of Upper Extremity (FMA-UE), Action Research Arm Test (ARAT), neurophysiological measures, and functional near-infrared spectroscopy (fNIRS) were assessed before (T0), at the midpoint (T1), and after the intervention (T2). Outcomes The patient exhibited post-intervention improvement in upper extremity function. There was increased excitability in the ipsilesional hemisphere and the opposite in the contralesional hemisphere. The interhemispheric inhibition (IHI) ratio increased from 2.70 to 10.81 and finally decreased to 1.34. Oxy-Hb signal was significantly decreased in affected M1 and mildly decreased in unaffected M1, while that of PMC and SMA on the affected side increased significantly. Conclusion The sequential inhibitory and facilitatory rTMS significantly promoted motor recovery in the patient. Related mechanisms include upregulation of excitability in the ipsilesional hemisphere, return of interhemispheric balance, and neuroplasticity-induced cortical reorganization.
Collapse
|
48
|
Zhang TR, Askari B, Kesici A, Guilherme E, Vila-Rodriguez F, Snyder JS. Intermittent theta burst transcranial magnetic stimulation induces hippocampal mossy fibre plasticity in male but not female mice. Eur J Neurosci 2023; 57:310-323. [PMID: 36484786 DOI: 10.1111/ejn.15891] [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: 06/02/2022] [Revised: 11/12/2022] [Accepted: 11/28/2022] [Indexed: 12/14/2022]
Abstract
Transcranial magnetic stimulation (TMS) induces electric fields that depolarise or hyperpolarise neurons. Intermittent theta burst stimulation (iTBS), a patterned form of TMS that is delivered at the theta frequency (~5 Hz), induces neuroplasticity in the hippocampus, a brain region that is implicated in memory and learning. One form of plasticity that is unique to the hippocampus is adult neurogenesis; however, little is known about whether TMS or iTBS in particular affects newborn neurons. Here, we therefore applied repeated sessions of iTBS to male and female mice and measured the extent of adult neurogenesis and the morphological features of immature neurons. We found that repeated sessions of iTBS did not significantly increase the amount of neurogenesis or affect the gross dendritic morphology of new neurons, and there were no sex differences in neurogenesis rates or aspects of afferent morphology. In contrast, efferent properties of newborn neurons varied as a function of sex and stimulation. Chronic iTBS increased the size of mossy fibre terminals, which synapse onto Cornu Ammonis 3 (CA3) pyramidal neurons, but only in males. iTBS also increased the number of terminal-associated filopodia, putative synapses onto inhibitory interneurons but only in male mice. This efferent plasticity could result from a general trophic effect, or it could reflect accelerated maturation of immature neurons. Given the important role of mossy fibre synapses in hippocampal learning, our results identify a neurobiological effect of iTBS that might be associated with sex-specific changes in cognition.
Collapse
Affiliation(s)
- Tian Rui Zhang
- Department of Psychology, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, British Columbia, Canada
- Non-Invasive Neurostimulation Therapies Laboratory, Department of Psychiatry, University of British Columbia, Vancouver, British Columbia, Canada
| | - Baran Askari
- Department of Psychology, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, British Columbia, Canada
| | - Aydan Kesici
- Department of Psychology, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, British Columbia, Canada
| | - Evelyn Guilherme
- Department of Physiotherapy, Federal University of Sao Carlos, Sao Carlo, Brazil
| | - Fidel Vila-Rodriguez
- Non-Invasive Neurostimulation Therapies Laboratory, Department of Psychiatry, University of British Columbia, Vancouver, British Columbia, Canada
| | - Jason S Snyder
- Department of Psychology, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
49
|
Jannati A, Oberman LM, Rotenberg A, Pascual-Leone A. Assessing the mechanisms of brain plasticity by transcranial magnetic stimulation. Neuropsychopharmacology 2023; 48:191-208. [PMID: 36198876 PMCID: PMC9700722 DOI: 10.1038/s41386-022-01453-8] [Citation(s) in RCA: 69] [Impact Index Per Article: 34.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 09/01/2022] [Accepted: 09/02/2022] [Indexed: 11/10/2022]
Abstract
Transcranial magnetic stimulation (TMS) is a non-invasive technique for focal brain stimulation based on electromagnetic induction where a fluctuating magnetic field induces a small intracranial electric current in the brain. For more than 35 years, TMS has shown promise in the diagnosis and treatment of neurological and psychiatric disorders in adults. In this review, we provide a brief introduction to the TMS technique with a focus on repetitive TMS (rTMS) protocols, particularly theta-burst stimulation (TBS), and relevant rTMS-derived metrics of brain plasticity. We then discuss the TMS-EEG technique, the use of neuronavigation in TMS, the neural substrate of TBS measures of plasticity, the inter- and intraindividual variability of those measures, effects of age and genetic factors on TBS aftereffects, and then summarize alterations of TMS-TBS measures of plasticity in major neurological and psychiatric disorders including autism spectrum disorder, schizophrenia, depression, traumatic brain injury, Alzheimer's disease, and diabetes. Finally, we discuss the translational studies of TMS-TBS measures of plasticity and their therapeutic implications.
Collapse
Affiliation(s)
- Ali Jannati
- Neuromodulation Program, Division of Epilepsy and Clinical Neurophysiology, Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA.
- F. M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA.
- Berenson-Allen Center for Noninvasive Brain Stimulation, Division of Cognitive Neurology, Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA.
| | - Lindsay M Oberman
- Center for Neuroscience and Regenerative Medicine, Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA
| | - Alexander Rotenberg
- Neuromodulation Program, Division of Epilepsy and Clinical Neurophysiology, Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
- F. M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
- Berenson-Allen Center for Noninvasive Brain Stimulation, Division of Cognitive Neurology, Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Alvaro Pascual-Leone
- Department of Neurology, Harvard Medical School, Boston, MA, USA.
- Hinda and Arthur Marcus Institute for Aging Research and Deanna and Sidney Wolk Center for Memory Health, Hebrew SeniorLife, Boston, MA, USA.
- Guttmann Brain Health Institute, Institut Guttmann, Barcelona, Spain.
| |
Collapse
|
50
|
Kweon J, Vigne MM, Jones RN, Carpenter LL, Brown JC. Practice makes plasticity: 10-Hz rTMS enhances LTP-like plasticity in musicians and athletes. Front Neural Circuits 2023; 17:1124221. [PMID: 37025991 PMCID: PMC10070804 DOI: 10.3389/fncir.2023.1124221] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 02/24/2023] [Indexed: 04/08/2023] Open
Abstract
Motor skill learning has been linked to functional and structural changes in the brain. Musicians and athletes undergo intensive motor training through the practice of an instrument or sport and have demonstrated use-dependent plasticity that may be subserved by long-term potentiation (LTP) processes. We know less, however, about whether the brains of musicians and athletes respond to plasticity-inducing interventions, such as repetitive transcranial magnetic stimulation (rTMS), differently than those without extensive motor training. In a pharmaco-rTMS study, we evaluated motor cortex excitability before and after an rTMS protocol in combination with oral administration of D-cycloserine (DCS) or placebo. In a secondary covariate analysis, we compared results between self-identified musicians and athletes (M&As) and non-musicians and athletes (non-M&As). Three TMS measures of cortical physiology were used to evaluate plasticity. We found that M&As did not have higher baseline corticomotor excitability. However, a plasticity-inducing protocol (10-Hz rTMS in combination with DCS) strongly facilitated motor-evoked potentials (MEPs) in M&As, but only weakly in non-M&As. Placebo and rTMS produced modest facilitation in both groups. Our findings suggest that motor practice and learning create a neuronal environment more responsive to plasticity-inducing events, including rTMS. These findings may explain one factor contributing to the high inter-individual variability found with MEP data. Greater capacity for plasticity holds implications for learning paradigms, such as psychotherapy and rehabilitation, by facilitating LTP-like activation of key networks, including recovery from neurological/mental disorders.
Collapse
Affiliation(s)
- Jamie Kweon
- Neuromodulation Research Facility, TMS Clinic, Butler Hospital, Providence, RI, United States
| | - Megan M. Vigne
- Neuromodulation Research Facility, TMS Clinic, Butler Hospital, Providence, RI, United States
| | - Richard N. Jones
- Department of Psychiatry and Human Behavior, Warren Alpert Medical School of Brown University, Providence, RI, United States
- Department of Neurology, Warren Alpert Medical School of Brown University, Providence, RI, United States
| | - Linda L. Carpenter
- Neuromodulation Research Facility, TMS Clinic, Butler Hospital, Providence, RI, United States
- Department of Psychiatry and Human Behavior, Warren Alpert Medical School of Brown University, Providence, RI, United States
| | - Joshua C. Brown
- Neuromodulation Research Facility, TMS Clinic, Butler Hospital, Providence, RI, United States
- Department of Psychiatry and Human Behavior, Warren Alpert Medical School of Brown University, Providence, RI, United States
- Department of Neurology, Warren Alpert Medical School of Brown University, Providence, RI, United States
- *Correspondence: Joshua C. Brown
| |
Collapse
|