1
|
Fischer QS, Kalikulov D, Viana Di Prisco G, Williams CA, Baldwin PR, Friedlander MJ. Synaptic Plasticity in the Injured Brain Depends on the Temporal Pattern of Stimulation. J Neurotrauma 2024; 41:2455-2477. [PMID: 38818799 DOI: 10.1089/neu.2024.0129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/01/2024] Open
Abstract
Neurostimulation protocols are increasingly used as therapeutic interventions, including for brain injury. In addition to the direct activation of neurons, these stimulation protocols are also likely to have downstream effects on those neurons' synaptic outputs. It is well known that alterations in the strength of synaptic connections (long-term potentiation, LTP; long-term depression, LTD) are sensitive to the frequency of stimulation used for induction; however, little is known about the contribution of the temporal pattern of stimulation to the downstream synaptic plasticity that may be induced by neurostimulation in the injured brain. We explored interactions of the temporal pattern and frequency of neurostimulation in the normal cerebral cortex and after mild traumatic brain injury (mTBI), to inform therapies to strengthen or weaken neural circuits in injured brains, as well as to better understand the role of these factors in normal brain plasticity. Whole-cell (WC) patch-clamp recordings of evoked postsynaptic potentials in individual neurons, as well as field potential (FP) recordings, were made from layer 2/3 of visual cortex in response to stimulation of layer 4, in acute slices from control (naive), sham operated, and mTBI rats. We compared synaptic plasticity induced by different stimulation protocols, each consisting of a specific frequency (1 Hz, 10 Hz, or 100 Hz), continuity (continuous or discontinuous), and temporal pattern (perfectly regular, slightly irregular, or highly irregular). At the individual neuron level, dramatic differences in plasticity outcome occurred when the highly irregular stimulation protocol was used at 1 Hz or 10 Hz, producing an overall LTD in controls and shams, but a robust overall LTP after mTBI. Consistent with the individual neuron results, the plasticity outcomes for simultaneous FP recordings were similar, indicative of our results generalizing to a larger scale synaptic network than can be sampled by individual WC recordings alone. In addition to the differences in plasticity outcome between control (naive or sham) and injured brains, the dynamics of the changes in synaptic responses that developed during stimulation were predictive of the final plasticity outcome. Our results demonstrate that the temporal pattern of stimulation plays a role in the polarity and magnitude of synaptic plasticity induced in the cerebral cortex while highlighting differences between normal and injured brain responses. Moreover, these results may be useful for optimization of neurostimulation therapies to treat mTBI and other brain disorders, in addition to providing new insights into downstream plasticity signaling mechanisms in the normal brain.
Collapse
Affiliation(s)
- Quentin S Fischer
- Fralin Biomedical Research Institute at VTC, Roanoke, Virginia, USA
- FBRI Center for Neurobiology Research, Roanoke, Virginia, USA
- Department of Neuroscience, Baylor College of Medicine, Houston, Texas, USA
| | - Djanenkhodja Kalikulov
- Fralin Biomedical Research Institute at VTC, Roanoke, Virginia, USA
- FBRI Center for Neurobiology Research, Roanoke, Virginia, USA
- Department of Neuroscience, Baylor College of Medicine, Houston, Texas, USA
| | | | - Carrie A Williams
- Fralin Biomedical Research Institute at VTC, Roanoke, Virginia, USA
- FBRI Center for Neurobiology Research, Roanoke, Virginia, USA
| | - Philip R Baldwin
- Department of Neuroscience, Baylor College of Medicine, Houston, Texas, USA
| | - Michael J Friedlander
- Fralin Biomedical Research Institute at VTC, Roanoke, Virginia, USA
- FBRI Center for Neurobiology Research, Roanoke, Virginia, USA
- Department of Neuroscience, Baylor College of Medicine, Houston, Texas, USA
- Department of Biological Sciences, Virginia Tech, Blacksburg, Virginia, USA
- Department of Psychiatry and Behavioral Medicine, Virginia Tech Carilion School of Medicine, Roanoke, Virginia, USA
- Faculty of Health Sciences, Virginia Tech, Roanoke, Virginia, USA
| |
Collapse
|
2
|
Qian F, He R, Du X, Wei Y, Zhou Z, Fan J, He Y. Microglia and Astrocytes Responses Contribute to Alleviating Inflammatory Damage by Repetitive Transcranial Magnetic Stimulation in Rats with Traumatic Brain Injury. Neurochem Res 2024; 49:2636-2651. [PMID: 38909329 DOI: 10.1007/s11064-024-04197-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 05/30/2024] [Accepted: 06/14/2024] [Indexed: 06/24/2024]
Abstract
Repetitive transcranial magnetic stimulation (rTMS) is a therapeutic strategy that shows promise in ameliorating the clinical sequelae following traumatic brain injury (TBI). These improvements are associated with neuroplastic changes in neurons and their synaptic connections. However, it has been hypothesized that rTMS may also modulate microglia and astrocytes, potentially potentiating their neuroprotective capabilities. This study aims to investigate the effects of high-frequency rTMS on microglia and astrocytes that may contribute to its neuroprotective effects. Feeney's weight-dropping method was used to establish rat models of moderate TBI. To evaluate the neuroprotective effect of high frequency rTMS on rats by observing the synaptic ultrastructure and the level of neuron apoptosis. The levels of several important inflammation-related proteins within microglia and astrocytes were assessed through immunofluorescence staining and western blot. Our findings demonstrate that injured neurons can be rescued through the modulation of microglia and astrocytes by rTMS. This modulation plays a key role in preserving the synaptic ultrastructure and inhibiting neuronal apoptosis. Among microglia, we observed that rTMS inhibited the levels of proinflammatory factors (CD16, IL-6 and TNF-α) and promoted the levels of anti-inflammatory factors (CD206, IL-10 and TNF-β). rTMS also reduced the levels of pyroptosis within microglia and pyroptosis-related proteins (NLRP3, Caspase-1, GSDMD, IL-1β and IL-18). Moreover, rTMS downregulated P75NTR expression and up-regulated IL33 expression in astrocytes. These findings suggest that regulation of microglia and astrocytes is the mechanism through which rTMS attenuates neuronal inflammatory damage after moderate TBI.
Collapse
Affiliation(s)
- FangFang Qian
- Department of Rehabilitation Medicine, Guangdong Province, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Avenue, Guangzhou, 510515, China
| | - RenHong He
- Department of Rehabilitation Medicine, Guangdong Province, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Avenue, Guangzhou, 510515, China
| | - XiaoHui Du
- Department of Rehabilitation Medicine, Guangdong Province, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Avenue, Guangzhou, 510515, China
| | - Yi Wei
- Department of Rehabilitation Medicine, Guangdong Province, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Avenue, Guangzhou, 510515, China
| | - Zhou Zhou
- Department of Rehabilitation Medicine, Guangdong Province, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Avenue, Guangzhou, 510515, China
| | - JianZhong Fan
- Department of Rehabilitation Medicine, Guangdong Province, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Avenue, Guangzhou, 510515, China.
| | - YouHua He
- Department of Comprehensive Medical Treatment Ward, Guangdong Province, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Avenue, Guangzhou, 510515, China.
| |
Collapse
|
3
|
McGregor KM, Novak T, Nocera JR, Mammino K, Wolf SL, Krishnamurthy LC. Examination of acute spin exercise on GABA levels in aging and stroke: The EASE study protocol. PLoS One 2024; 19:e0297841. [PMID: 39008457 PMCID: PMC11249249 DOI: 10.1371/journal.pone.0297841] [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: 05/29/2023] [Accepted: 01/08/2024] [Indexed: 07/17/2024] Open
Abstract
BACKGROUND Changes in regional levels of the inhibitory neurotransmitter gamma-aminobutyric acid (GABA) may indicate the potential for favorable responses to the treatment of stroke affecting the upper extremity. By selectively altering GABA levels during training, we may induce long-term potentiation and adjust excitatory/inhibitory balance (E/I balance). However, the impact of this alteration may be limited by neural damage or aging. Aerobic exercise has been shown to increase GABA levels in the sensorimotor cortex and improve motor learning by widening the dynamic range of E/I balance. The cross-sectional project, Effects of Acute Exercise on Functional Magnetic Resonance Spectroscopy Measures of GABA in Aging and Chronic Stroke (EASE), is designed to assess the functional relevance of changes in GABA concentration within the sensorimotor cortex before and after an acute aerobic exercise session. METHODS/DESIGN EASE will enroll 30 participants comprised of healthy younger adults (18-35 years; n = 10), older adults (60+ years; n = 10), and persons with chronic stroke (n = 10) affecting distal upper extremity function. We will use resting magnetic resonance spectroscopy to measure all participants' GABA levels at rest before and after aerobic exercise. In addition, we will employ functional magnetic resonance spectroscopy using motor skill acquisition and recall tasks in healthy adults. We hypothesize that acute aerobic exercise will increase resting sensorimotor GABA concentration and that higher GABA resting levels will predict better motor learning performance on measures taken both inside and outside the magnet. We also hypothesize that a higher dynamic range of GABA during task-based spectroscopy in healthy adults will predict better motor skill acquisition and recall. DISCUSSION The EASE project will evaluate the effect of acute exercise on GABA levels as a biomarker of upper extremity motor skill learning with two populations (aging adults and those with chronic stroke). We predict that acute exercise, higher sensorimotor GABA levels, and broader dynamic range will be related to better motor skill acquisition.
Collapse
Affiliation(s)
- Keith M. McGregor
- Birmingham VA Geriatric Research Education and Clinical Center, Birmingham VA Health Care System, Birmingham, Alabama, United States of America
- Department of Clinical and Diagnostic Sciences, University of Alabama at Birmingham School of Health Professions, Birmingham, Alabama, United States of America
| | - Thomas Novak
- Rehabilitation R&D Center for Visual and Neurocognitive Rehabilitation, Atlanta VA Health Care System, Atlanta, Georgia, United States of America
- Department of Neurology, Emory University School of Medicine, Atlanta, Georgia, United States of America
- Division of Physical Therapy, Department of Rehabilitation Medicine, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Joe R. Nocera
- Rehabilitation R&D Center for Visual and Neurocognitive Rehabilitation, Atlanta VA Health Care System, Atlanta, Georgia, United States of America
- Department of Neurology, Emory University School of Medicine, Atlanta, Georgia, United States of America
- Division of Physical Therapy, Department of Rehabilitation Medicine, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Kevin Mammino
- Rehabilitation R&D Center for Visual and Neurocognitive Rehabilitation, Atlanta VA Health Care System, Atlanta, Georgia, United States of America
| | - Steven L. Wolf
- Rehabilitation R&D Center for Visual and Neurocognitive Rehabilitation, Atlanta VA Health Care System, Atlanta, Georgia, United States of America
- Division of Physical Therapy, Department of Rehabilitation Medicine, Emory University School of Medicine, Atlanta, Georgia, United States of America
- Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, United States of America
- Department of Cell Biology, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Lisa C. Krishnamurthy
- Atlanta VA Health Care System, Decatur, Georgia, United States of America
- Tri-Institutional Center for Translational Research in Neuroimaging and Data Science (TReNDS), Georgia State, Georgia Tech and Emory, Atlanta, Georgia, United States of America
- Department of Physics & Astronomy, Georgia State University, Atlanta, Georgia, United States of America
- Department of Radiology and Imaging Sciences, Emory University, Atlanta, Georgia, United States of America
| |
Collapse
|
4
|
Revelo Herrera SG, Leon-Rojas JE. The Effect of Aerobic Exercise in Neuroplasticity, Learning, and Cognition: A Systematic Review. Cureus 2024; 16:e54021. [PMID: 38476815 PMCID: PMC10932589 DOI: 10.7759/cureus.54021] [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: 02/11/2024] [Indexed: 03/14/2024] Open
Abstract
This systematic review aims to examine the association between physical activity, neuroplasticity, and cognition. We analyzed an initial dataset consisting of 9935 articles retrieved from three scientific platforms (PubMed, Scopus, and the Virtual Health Library). Various screening filters were applied to refine the information against predefined eligibility criteria, resulting in the inclusion of a total of 17 articles that assessed the effect of aerobic exercise on neuroplasticity. The results suggested that aerobic exercise at various intensities, particularly at high intensity, can influence cortical excitability and result in cognitive improvement; also, exercise was associated with direct cortical and structural changes. Exercise has shown efficacy in individuals of diverse age groups, as well as in people with and without brain disease.
Collapse
|
5
|
Nie L, He J, Wang J, Wang R, Huang L, Jia L, Kim YT, Bhawal UK, Fan X, Zille M, Jiang C, Chen X, Wang J. Environmental Enrichment for Stroke and Traumatic Brain Injury: Mechanisms and Translational Implications. Compr Physiol 2023; 14:5291-5323. [PMID: 38158368 DOI: 10.1002/cphy.c230007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2024]
Abstract
Acquired brain injuries, such as ischemic stroke, intracerebral hemorrhage (ICH), and traumatic brain injury (TBI), can cause severe neurologic damage and even death. Unfortunately, currently, there are no effective and safe treatments to reduce the high disability and mortality rates associated with these brain injuries. However, environmental enrichment (EE) is an emerging approach to treating and rehabilitating acquired brain injuries by promoting motor, sensory, and social stimulation. Multiple preclinical studies have shown that EE benefits functional recovery, including improved motor and cognitive function and psychological benefits mediated by complex protective signaling pathways. This article provides an overview of the enriched environment protocols used in animal models of ischemic stroke, ICH, and TBI, as well as relevant clinical studies, with a particular focus on ischemic stroke. Additionally, we explored studies of animals with stroke and TBI exposed to EE alone or in combination with multiple drugs and other rehabilitation modalities. Finally, we discuss the potential clinical applications of EE in future brain rehabilitation therapy and the molecular and cellular changes caused by EE in rodents with stroke or TBI. This article aims to advance preclinical and clinical research on EE rehabilitation therapy for acquired brain injury. © 2024 American Physiological Society. Compr Physiol 14:5291-5323, 2024.
Collapse
Affiliation(s)
- Luwei Nie
- Department of Pain Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan Province, China
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jinxin He
- Department of Pain Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan Province, China
- Key Laboratory for Brain Science Research and Transformation in the Tropical Environment of Hainan Province, Hainan Medical University, Haikou, China
| | - Junmin Wang
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan Province, China
| | - Ruike Wang
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan Province, China
| | - Leo Huang
- Department of Psychology, University of Toronto, Toronto, Ontario, Canada
| | - Lin Jia
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan Province, China
| | - Yun Tai Kim
- Division of Functional Food Research, Korea Food Research Institute, Wanju-gun, Jeollabuk-do, Republic of Korea
- Department of Food Biotechnology, Korea University of Science & Technology, Daejeon, Republic of Korea
| | - Ujjal K Bhawal
- Research Institute of Oral Science, Nihon University School of Dentistry at Matsudo, Chiba, Japan
- Center for Global Health Research, Saveetha Medical College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, Tamil Nadu, India
| | - Xiaochong Fan
- Department of Pain Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Marietta Zille
- Department of Pharmaceutical Sciences, Division of Pharmacology and Toxicology, University of Vienna, Vienna, Austria
| | - Chao Jiang
- Department of Neurology, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| | - Xuemei Chen
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan Province, China
| | - Jian Wang
- Department of Pain Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan Province, China
| |
Collapse
|
6
|
Bozkurt S, Lannin NA, Mychasiuk R, Semple BD. Environmental modifications to rehabilitate social behavior deficits after acquired brain injury: What is the evidence? Neurosci Biobehav Rev 2023; 152:105278. [PMID: 37295762 DOI: 10.1016/j.neubiorev.2023.105278] [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: 01/18/2023] [Revised: 04/22/2023] [Accepted: 05/20/2023] [Indexed: 06/12/2023]
Abstract
Social behavior deficits are a common, debilitating consequence of traumatic brain injury and stroke, particularly when sustained during childhood. Numerous factors influence the manifestation of social problems after acquired brain injuries, raising the question of whether environmental manipulations can minimize or prevent such deficits. Here, we examine both clinical and preclinical evidence addressing this question, with a particular focus on environmental enrichment paradigms and differing housing conditions. We aimed to understand whether environmental manipulations can ameliorate injury-induced social behavior deficits. In summary, promising data from experimental models supports a beneficial role of environmental enrichment on social behavior. However, limited studies have considered social outcomes in the chronic setting, and few studies have addressed the social context specifically as an important component of the post-injury environment. Clinically, limited high-caliber evidence supports the use of specific interventions for social deficits after acquired brain injuries. An improved understanding of how the post-injury environment interacts with the injured brain, particularly during development, is needed to validate the implementation of rehabilitative interventions that involve manipulating an individuals' environment.
Collapse
Affiliation(s)
- Salome Bozkurt
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Natasha A Lannin
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia; Alfred Health, Melbourne, VIC, Australia; School of Allied Health (Occupational Therapy), La Trobe University, Melbourne, Australia
| | - Richelle Mychasiuk
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia; Alfred Health, Melbourne, VIC, Australia
| | - Bridgette D Semple
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia; Alfred Health, Melbourne, VIC, Australia; Department of Medicine (Royal Melbourne Hospital), The University of Melbourne, Parkville, VIC, Australia.
| |
Collapse
|
7
|
Netzley AH, Pelled G. The Pig as a Translational Animal Model for Biobehavioral and Neurotrauma Research. Biomedicines 2023; 11:2165. [PMID: 37626662 PMCID: PMC10452425 DOI: 10.3390/biomedicines11082165] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 07/27/2023] [Accepted: 07/28/2023] [Indexed: 08/27/2023] Open
Abstract
In recent decades, the pig has attracted considerable attention as an important intermediary model animal in translational biobehavioral research due to major similarities between pig and human neuroanatomy, physiology, and behavior. As a result, there is growing interest in using pigs to model many human neurological conditions and injuries. Pigs are highly intelligent and are capable of performing a wide range of behaviors, which can provide valuable insight into the effects of various neurological disease states. One area in which the pig has emerged as a particularly relevant model species is in the realm of neurotrauma research. Indeed, the number of investigators developing injury models and assessing treatment options in pigs is ever-expanding. In this review, we examine the use of pigs for cognitive and behavioral research as well as some commonly used physiological assessment methods. We also discuss the current usage of pigs as a model for the study of traumatic brain injury. We conclude that the pig is a valuable animal species for studying cognition and the physiological effect of disease, and it has the potential to contribute to the development of new treatments and therapies for human neurological and psychiatric disorders.
Collapse
Affiliation(s)
- Alesa H. Netzley
- Department of Biomedical Engineering, Michigan State University, East Lansing, MI 48824, USA;
| | - Galit Pelled
- Neuroscience Program, Michigan State University, East Lansing, MI 48824, USA
- Department of Mechanical Engineering, Michigan State University, East Lansing, MI 48824, USA
- Department of Radiology, Michigan State University, East Lansing, MI 48824, USA
| |
Collapse
|
8
|
Evancho A, Tyler WJ, McGregor K. A review of combined neuromodulation and physical therapy interventions for enhanced neurorehabilitation. Front Hum Neurosci 2023; 17:1151218. [PMID: 37545593 PMCID: PMC10400781 DOI: 10.3389/fnhum.2023.1151218] [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: 01/25/2023] [Accepted: 06/30/2023] [Indexed: 08/08/2023] Open
Abstract
Rehabilitation approaches for individuals with neurologic conditions have increasingly shifted toward promoting neuroplasticity for enhanced recovery and restoration of function. This review focuses on exercise strategies and non-invasive neuromodulation techniques that target neuroplasticity, including transcranial magnetic stimulation (TMS), vagus nerve stimulation (VNS), and peripheral nerve stimulation (PNS). We have chosen to focus on non-invasive neuromodulation techniques due to their greater potential for integration into routine clinical practice. We explore and discuss the application of these interventional strategies in four neurological conditions that are frequently encountered in rehabilitation settings: Parkinson's Disease (PD), Traumatic Brain Injury (TBI), stroke, and Spinal Cord Injury (SCI). Additionally, we discuss the potential benefits of combining non-invasive neuromodulation with rehabilitation, which has shown promise in accelerating recovery. Our review identifies studies that demonstrate enhanced recovery through combined exercise and non-invasive neuromodulation in the selected patient populations. We primarily focus on the motor aspects of rehabilitation, but also briefly address non-motor impacts of these conditions. Additionally, we identify the gaps in current literature and barriers to implementation of combined approaches into clinical practice. We highlight areas needing further research and suggest avenues for future investigation, aiming to enhance the personalization of the unique neuroplastic responses associated with each condition. This review serves as a resource for rehabilitation professionals and researchers seeking a comprehensive understanding of neuroplastic exercise interventions and non-invasive neuromodulation techniques tailored for specific diseases and diagnoses.
Collapse
Affiliation(s)
- Alexandra Evancho
- Department of Physical Therapy, School of Health Professions, University of Alabama at Birmingham, Birmingham, AL, United States
| | - William J. Tyler
- Department of Biomedical Engineering, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
- Department of Physical Medicine and Rehabilitation, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Keith McGregor
- Department of Clinical and Diagnostic Studies, School of Health Professions, University of Alabama at Birmingham, Birmingham, AL, United States
| |
Collapse
|
9
|
Ziesel D, Nowakowska M, Scheruebel S, Kornmueller K, Schäfer U, Schindl R, Baumgartner C, Üçal M, Rienmüller T. Electrical stimulation methods and protocols for the treatment of traumatic brain injury: a critical review of preclinical research. J Neuroeng Rehabil 2023; 20:51. [PMID: 37098582 PMCID: PMC10131365 DOI: 10.1186/s12984-023-01159-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Accepted: 03/13/2023] [Indexed: 04/27/2023] Open
Abstract
BACKGROUND Traumatic brain injury (TBI) is a leading cause of disabilities resulting from cognitive and neurological deficits, as well as psychological disorders. Only recently, preclinical research on electrical stimulation methods as a potential treatment of TBI sequelae has gained more traction. However, the underlying mechanisms of the anticipated improvements induced by these methods are still not fully understood. It remains unclear in which stage after TBI they are best applied to optimize the therapeutic outcome, preferably with persisting effects. Studies with animal models address these questions and investigate beneficial long- and short-term changes mediated by these novel modalities. METHODS In this review, we present the state-of-the-art in preclinical research on electrical stimulation methods used to treat TBI sequelae. We analyze publications on the most commonly used electrical stimulation methods, namely transcranial magnetic stimulation (TMS), transcranial direct current stimulation (tDCS), deep brain stimulation (DBS) and vagus nerve stimulation (VNS), that aim to treat disabilities caused by TBI. We discuss applied stimulation parameters, such as the amplitude, frequency, and length of stimulation, as well as stimulation time frames, specifically the onset of stimulation, how often stimulation sessions were repeated and the total length of the treatment. These parameters are then analyzed in the context of injury severity, the disability under investigation and the stimulated location, and the resulting therapeutic effects are compared. We provide a comprehensive and critical review and discuss directions for future research. RESULTS AND CONCLUSION: We find that the parameters used in studies on each of these stimulation methods vary widely, making it difficult to draw direct comparisons between stimulation protocols and therapeutic outcome. Persisting beneficial effects and adverse consequences of electrical simulation are rarely investigated, leaving many questions about their suitability for clinical applications. Nevertheless, we conclude that the stimulation methods discussed here show promising results that could be further supported by additional research in this field.
Collapse
Affiliation(s)
- D Ziesel
- Institute of Health Care Engineering with European Testing Center of Medical Devices, Graz University of Technology, Graz, Austria
| | - M Nowakowska
- Research Unit of Experimental Neurotraumatology, Department of Neurosurgery, Medical University of Graz, Graz, Austria
| | - S Scheruebel
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Biophysics Division, Medical University of Graz, Graz, Austria
| | - K Kornmueller
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Biophysics Division, Medical University of Graz, Graz, Austria
| | - U Schäfer
- Research Unit of Experimental Neurotraumatology, Department of Neurosurgery, Medical University of Graz, Graz, Austria
- BioTechMed-Graz, Graz, Austria
| | - R Schindl
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Biophysics Division, Medical University of Graz, Graz, Austria
- BioTechMed-Graz, Graz, Austria
| | - C Baumgartner
- Institute of Health Care Engineering with European Testing Center of Medical Devices, Graz University of Technology, Graz, Austria
- BioTechMed-Graz, Graz, Austria
| | - M Üçal
- Research Unit of Experimental Neurotraumatology, Department of Neurosurgery, Medical University of Graz, Graz, Austria
- BioTechMed-Graz, Graz, Austria
| | - T Rienmüller
- Institute of Health Care Engineering with European Testing Center of Medical Devices, Graz University of Technology, Graz, Austria.
- BioTechMed-Graz, Graz, Austria.
| |
Collapse
|
10
|
Lynch DG, Narayan RK, Li C. Multi-Mechanistic Approaches to the Treatment of Traumatic Brain Injury: A Review. J Clin Med 2023; 12:jcm12062179. [PMID: 36983181 PMCID: PMC10052098 DOI: 10.3390/jcm12062179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 03/08/2023] [Accepted: 03/09/2023] [Indexed: 03/18/2023] Open
Abstract
Traumatic brain injury (TBI) is a leading cause of death and disability worldwide. Despite extensive research efforts, the majority of trialed monotherapies to date have failed to demonstrate significant benefit. It has been suggested that this is due to the complex pathophysiology of TBI, which may possibly be addressed by a combination of therapeutic interventions. In this article, we have reviewed combinations of different pharmacologic treatments, combinations of non-pharmacologic interventions, and combined pharmacologic and non-pharmacologic interventions for TBI. Both preclinical and clinical studies have been included. While promising results have been found in animal models, clinical trials of combination therapies have not yet shown clear benefit. This may possibly be due to their application without consideration of the evolving pathophysiology of TBI. Improvements of this paradigm may come from novel interventions guided by multimodal neuromonitoring and multimodal imaging techniques, as well as the application of multi-targeted non-pharmacologic and endogenous therapies. There also needs to be a greater representation of female subjects in preclinical and clinical studies.
Collapse
Affiliation(s)
- Daniel G. Lynch
- Translational Brain Research Laboratory, The Feinstein Institutes for Medical Research, Manhasset, NY 11030, USA
- Zucker School of Medicine at Hofstra/Northwell Health, Hempstead, NY 11549, USA
| | - Raj K. Narayan
- Translational Brain Research Laboratory, The Feinstein Institutes for Medical Research, Manhasset, NY 11030, USA
- Department of Neurosurgery, St. Francis Hospital, Roslyn, NY 11576, USA
| | - Chunyan Li
- Translational Brain Research Laboratory, The Feinstein Institutes for Medical Research, Manhasset, NY 11030, USA
- Zucker School of Medicine at Hofstra/Northwell Health, Hempstead, NY 11549, USA
- Department of Neurosurgery, Northwell Health, Manhasset, NY 11030, USA
- Correspondence:
| |
Collapse
|
11
|
Surendrakumar S, Rabelo TK, Campos ACP, Mollica A, Abrahao A, Lipsman N, Burke MJ, Hamani C. Neuromodulation Therapies in Pre-Clinical Models of Traumatic Brain Injury: Systematic Review and Translational Applications. J Neurotrauma 2023; 40:435-448. [PMID: 35983592 DOI: 10.1089/neu.2022.0286] [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] [Indexed: 11/13/2022] Open
Abstract
Traumatic brain injury (TBI) has been associated with several lasting impairments that affect quality of life. Pre-clinical models of TBI have been studied to further our understanding of the underlying short-term and long-term symptomatology. Neuromodulation techniques have become of great interest in recent years as potential rehabilitative therapies after injury because of their capacity to alter neuronal activity and neural circuits in targeted brain regions. This systematic review aims to provide an overlook of the behavioral and neurochemical effects of transcranial direct current stimulation (tDCS), transcranial magnetic stimulation (TMS), deep brain stimulation (DBS), and vagus nerve stimulation (VNS) in pre-clinical TBI models. After screening 629 abstracts, 30 articles were pooled for review. These studies showed that tDCS, TMS, DBS, or VNS delivered to rodents restored TBI-induced deficits in coordination, balance, locomotor activity and improved cognitive impairments in memory, learning, and impulsivity. Potential mechanisms for these effects included neuroprotection, a decrease in apoptosis, neuroplasticity, and the restoration of neural circuit abnormalities. The translational value, potential applicability, and the interpretation of these findings in light of outcome data from clinical trials in patients with TBI are discussed.
Collapse
Affiliation(s)
- Shanan Surendrakumar
- Harquail Centre for Neuromodulation, Sunnybrook Health Sciences Centre, Sunnybrook Research Institute, Toronto, Ontario, Canada
| | - Thallita Kelly Rabelo
- Harquail Centre for Neuromodulation, Sunnybrook Health Sciences Centre, Sunnybrook Research Institute, Toronto, Ontario, Canada
| | - Ana Carolina P Campos
- Harquail Centre for Neuromodulation, Sunnybrook Health Sciences Centre, Sunnybrook Research Institute, Toronto, Ontario, Canada
| | - Adriano Mollica
- Harquail Centre for Neuromodulation, Sunnybrook Health Sciences Centre, Sunnybrook Research Institute, Toronto, Ontario, Canada.,Neuropsychiatry Program, Department of Psychiatry, Department of Medicine, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, Ontario, Canada
| | - Agessandro Abrahao
- Harquail Centre for Neuromodulation, Sunnybrook Health Sciences Centre, Sunnybrook Research Institute, Toronto, Ontario, Canada.,Hurvitz Brain Sciences Centre, Sunnybrook Research Institute, Toronto, Ontario, Canada.,Division of Neurology, Department of Medicine, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, Ontario, Canada
| | - Nir Lipsman
- Harquail Centre for Neuromodulation, Sunnybrook Health Sciences Centre, Sunnybrook Research Institute, Toronto, Ontario, Canada.,Hurvitz Brain Sciences Centre, Sunnybrook Research Institute, Toronto, Ontario, Canada.,Division of Neurology, Department of Medicine, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, Ontario, Canada
| | - Matthew J Burke
- Harquail Centre for Neuromodulation, Sunnybrook Health Sciences Centre, Sunnybrook Research Institute, Toronto, Ontario, Canada.,Hurvitz Brain Sciences Centre, Sunnybrook Research Institute, Toronto, Ontario, Canada.,Neuropsychiatry Program, Department of Psychiatry, Department of Medicine, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, Ontario, Canada
| | - Clement Hamani
- Harquail Centre for Neuromodulation, Sunnybrook Health Sciences Centre, Sunnybrook Research Institute, Toronto, Ontario, Canada.,Hurvitz Brain Sciences Centre, Sunnybrook Research Institute, Toronto, Ontario, Canada.,Division of Neurology, Department of Medicine, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
12
|
Wang Y, Li Q, Xue X, Xu X, Tao W, Liu S, Li Y, Wang H, Hua Y. Neuroplasticity of pain processing and motor control in CAI patients: A UK Biobank study with clinical validation. Front Mol Neurosci 2023; 16:1096930. [PMID: 36866356 PMCID: PMC9971622 DOI: 10.3389/fnmol.2023.1096930] [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: 11/13/2022] [Accepted: 01/16/2023] [Indexed: 02/16/2023] Open
Abstract
Background Pain plays an important role in chronic ankle instability (CAI), and prolonged pain may be associated with ankle dysfunction and abnormal neuroplasticity. Purpose To investigate the differences in resting-state functional connectivity among the pain-related brain regions and the ankle motor-related brain regions between healthy controls and patients with CAI, and explore the relationship between patients' motor function and pain. Study design A cross-database, cross-sectional study. Methods This study included a UK Biobank dataset of 28 patients with ankle pain and 109 healthy controls and a validation dataset of 15 patients with CAI and 15 healthy controls. All participants underwent resting-state functional magnetic resonance imaging scanning, and the functional connectivity (FC) among the pain-related brain regions and the ankle motor-related brain regions were calculated and compared between groups. The correlations between the potentially different functional connectivity and the clinical questionnaires were also explored in patients with CAI. Results The functional connection between the cingulate motor area and insula significantly differed between groups in both the UK Biobank (p = 0.005) and clinical validation dataset (p = 0.049), which was also significantly correlated with Tegner scores (r = 0.532, p = 0.041) in patients with CAI. Conclusion A reduced functional connection between the cingulate motor area and the insula was present in patients with CAI, which was also directly correlated with reduction in the level of patient physical activity.
Collapse
Affiliation(s)
- Yiran Wang
- Department of Sports Medicine, Fudan University, Shanghai, China
| | - Qianru Li
- Department of Sports Medicine, Fudan University, Shanghai, China
| | - Xiao'ao Xue
- Department of Sports Medicine, Fudan University, Shanghai, China
| | - Xiaoyun Xu
- School of Exercise and Health, Shanghai University of Sport, Shanghai, China
| | - Weichu Tao
- School of Exercise and Health, Shanghai University of Sport, Shanghai, China
| | - Sixu Liu
- Department of Biomedical Engineering, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yunyi Li
- Department of Biomedical Engineering, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - He Wang
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China,Human Phenome Institute, Fudan University, Shanghai, China,Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence, Ministry of Education, Fudan University, Shanghai, China,He Wang ✉
| | - Yinghui Hua
- Department of Sports Medicine, Fudan University, Shanghai, China,Yiwu Research Institute of Fudan University, Yiwu, China,*Correspondence: Yinghui Hua ✉
| |
Collapse
|
13
|
Wu T, Li M, Tian L, Cong P, Huang X, Wu H, Zhang Q, Zhang H, Xiong L. A modified mouse model of perioperative neurocognitive disorders exacerbated by sleep fragmentation. Exp Anim 2023; 72:55-67. [PMID: 36130912 PMCID: PMC9978123 DOI: 10.1538/expanim.22-0053] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
Aging is one of the greatest risk factors for postoperative cognitive dysfunction (POCD), also known as perioperative neurocognitive disorder (PND). Animal models of PND are usually induced in mice over 18 months of age, which imposes expensive economic and time costs for PND-related studies. Sleep disorders, including sleep fragmentation, are reported to aggravate memory impairment in neurocognitive-related diseases such as Alzheimer's disease (AD). Therefore, the aim of the present study was to explore whether a PND model could be constructed in younger mice with the help of fragmented sleep. We found that fragmented sleep followed by laparotomy under isoflurane anesthesia could stably induce PND in 15-month-old mice. To determine whether the neurocognitive decline in this model could be salvaged by clinical treatments, we administered repetitive transcranial magnetic stimulation (rTMS) to the model mice before anesthesia and surgery. We found that 10 days of high-frequency rTMS (HF-rTMS) could improve spatial learning and memory deficits in this modified PND model. We are the first to successfully construct a PND model in younger mice,which is more economical, that can be used as an alternative model for future PND studies.
Collapse
Affiliation(s)
- Tingmei Wu
- Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Department of
Anesthesiology and Perioperative Medicine, Shanghai Fourth People’s Hospital Affiliated to Tongji University School of Medicine, 1279 Sanmen Road, Hongkou District, Shanghai 200434, P.R.
China
| | - Min Li
- Department of Rehabilitation Medicine, Shanghai Fourth People’s Hospital Affiliated to Tongji University School of Medicine, 1279 Sanmen Road, Hongkou District,
Shanghai 200434, P.R. China
| | - Li Tian
- Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Department of
Anesthesiology and Perioperative Medicine, Shanghai Fourth People’s Hospital Affiliated to Tongji University School of Medicine, 1279 Sanmen Road, Hongkou District, Shanghai 200434, P.R.
China
| | - Peilin Cong
- Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Department of
Anesthesiology and Perioperative Medicine, Shanghai Fourth People’s Hospital Affiliated to Tongji University School of Medicine, 1279 Sanmen Road, Hongkou District, Shanghai 200434, P.R.
China
| | - Xinwei Huang
- Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Department of
Anesthesiology and Perioperative Medicine, Shanghai Fourth People’s Hospital Affiliated to Tongji University School of Medicine, 1279 Sanmen Road, Hongkou District, Shanghai 200434, P.R.
China
| | - Huanghui Wu
- Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Department of
Anesthesiology and Perioperative Medicine, Shanghai Fourth People’s Hospital Affiliated to Tongji University School of Medicine, 1279 Sanmen Road, Hongkou District, Shanghai 200434, P.R.
China
| | - Qian Zhang
- Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Department of
Anesthesiology and Perioperative Medicine, Shanghai Fourth People’s Hospital Affiliated to Tongji University School of Medicine, 1279 Sanmen Road, Hongkou District, Shanghai 200434, P.R.
China
| | - Hong Zhang
- Department of Rehabilitation Medicine, Shanghai Fourth People’s Hospital Affiliated to Tongji University School of Medicine, 1279 Sanmen Road, Hongkou District,
Shanghai 200434, P.R. China
| | - Lize Xiong
- Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Department of
Anesthesiology and Perioperative Medicine, Shanghai Fourth People’s Hospital Affiliated to Tongji University School of Medicine, 1279 Sanmen Road, Hongkou District, Shanghai 200434, P.R.
China
| |
Collapse
|
14
|
He RH, Fan JZ, Qian FF, He YH, Du XH, Lu HX. Repetitive transcranial magnetic stimulation promotes neurological functional recovery in rats with traumatic brain injury by upregulating synaptic plasticity-related proteins. Neural Regen Res 2023; 18:368-374. [PMID: 35900432 PMCID: PMC9396518 DOI: 10.4103/1673-5374.346548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Studies have shown that repetitive transcranial magnetic stimulation (rTMS) can enhance synaptic plasticity and improve neurological dysfunction. However, the mechanism through which rTMS can improve moderate traumatic brain injury remains poorly understood. In this study, we established rat models of moderate traumatic brain injury using Feeney’s weight-dropping method and treated them using rTMS. To help determine the mechanism of action, we measured levels of several important brain activity-related proteins and their mRNA. On the injured side of the brain, we found that rTMS increased the protein levels and mRNA expression of brain-derived neurotrophic factor, tropomyosin receptor kinase B, N-methyl-D-aspartic acid receptor 1, and phosphorylated cAMP response element binding protein, which are closely associated with the occurrence of long-term potentiation. rTMS also partially reversed the loss of synaptophysin after injury and promoted the remodeling of synaptic ultrastructure. These findings suggest that upregulation of synaptic plasticity-related protein expression is the mechanism through which rTMS promotes neurological function recovery after moderate traumatic brain injury.
Collapse
|
15
|
Lindsey A, Ellison RL, Herrold AA, Aaronson AL, Kletzel SL, Stika MM, Guernon A, Bender Pape T. rTMS/iTBS and Cognitive Rehabilitation for Deficits Associated With TBI and PTSD: A Theoretical Framework and Review. J Neuropsychiatry Clin Neurosci 2022; 35:28-38. [PMID: 35872613 DOI: 10.1176/appi.neuropsych.21090227] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Rehabilitation of cognitive and psychosocial deficits resulting from traumatic brain injury (TBI) continues to be an area of concern in health care. Commonly co-occurring psychiatric disorders, such as major depressive disorder and posttraumatic stress disorder, create additional hurdles when attempting to remediate cognitive sequelae. There is increased need for procedures that will yield consistent gains indicative of recovery of function. Intermittent theta-burst stimulation (iTBS), a form of repetitive transcranial magnetic stimulation, has potential as an instrument that can be tailored to aid cognitive processes and support functional gains. The use of iTBS enables direct stimulation of desired neural systems. iTBS, performed in conjunction with behavioral interventions (e.g., cognitive rehabilitation, psychotherapy), may result in additive success in facilitating cognitive restoration and adaptation. The purpose of this theoretical review is to illustrate how the technical and physiological aspects of iTBS may enhance other forms of neurorehabilitation for individuals with TBI. Future research on combinatorial iTBS interventions has the potential to translate to other complex neuropsychiatric conditions.
Collapse
Affiliation(s)
- André Lindsey
- Research Service (Lindsey, Ellison, Herrold, Kletzel, Guernon, Pape), Center for Innovation for Complex Chronic Healthcare (Herrold, aronson, Kletzel, Pape), and Spinal Cord Injury/Disorder Service (Stika), Edward Hines, Jr., Veterans Affairs (VA) Hospital, Hines, IL; School of Education, Nevada State College, Henderson (Lindsey); Department of Psychology, Illinois Institute of Technology, Chicago (Ellison); Departments of Psychiatry and Behavioral Medicine (Herrold, Aaronson) and Physical Medicine and Rehabilitation (Pape), Feinberg School of Medicine, Northwestern University, Chicago; Speech-Language Pathology Program, College of Nursing and Health Sciences, Lewis University, Romeoville, IL (Guernon)
| | - Rachael L Ellison
- Research Service (Lindsey, Ellison, Herrold, Kletzel, Guernon, Pape), Center for Innovation for Complex Chronic Healthcare (Herrold, aronson, Kletzel, Pape), and Spinal Cord Injury/Disorder Service (Stika), Edward Hines, Jr., Veterans Affairs (VA) Hospital, Hines, IL; School of Education, Nevada State College, Henderson (Lindsey); Department of Psychology, Illinois Institute of Technology, Chicago (Ellison); Departments of Psychiatry and Behavioral Medicine (Herrold, Aaronson) and Physical Medicine and Rehabilitation (Pape), Feinberg School of Medicine, Northwestern University, Chicago; Speech-Language Pathology Program, College of Nursing and Health Sciences, Lewis University, Romeoville, IL (Guernon)
| | - Amy A Herrold
- Research Service (Lindsey, Ellison, Herrold, Kletzel, Guernon, Pape), Center for Innovation for Complex Chronic Healthcare (Herrold, aronson, Kletzel, Pape), and Spinal Cord Injury/Disorder Service (Stika), Edward Hines, Jr., Veterans Affairs (VA) Hospital, Hines, IL; School of Education, Nevada State College, Henderson (Lindsey); Department of Psychology, Illinois Institute of Technology, Chicago (Ellison); Departments of Psychiatry and Behavioral Medicine (Herrold, Aaronson) and Physical Medicine and Rehabilitation (Pape), Feinberg School of Medicine, Northwestern University, Chicago; Speech-Language Pathology Program, College of Nursing and Health Sciences, Lewis University, Romeoville, IL (Guernon)
| | - Alexandra L Aaronson
- Research Service (Lindsey, Ellison, Herrold, Kletzel, Guernon, Pape), Center for Innovation for Complex Chronic Healthcare (Herrold, aronson, Kletzel, Pape), and Spinal Cord Injury/Disorder Service (Stika), Edward Hines, Jr., Veterans Affairs (VA) Hospital, Hines, IL; School of Education, Nevada State College, Henderson (Lindsey); Department of Psychology, Illinois Institute of Technology, Chicago (Ellison); Departments of Psychiatry and Behavioral Medicine (Herrold, Aaronson) and Physical Medicine and Rehabilitation (Pape), Feinberg School of Medicine, Northwestern University, Chicago; Speech-Language Pathology Program, College of Nursing and Health Sciences, Lewis University, Romeoville, IL (Guernon)
| | - Sandra L Kletzel
- Research Service (Lindsey, Ellison, Herrold, Kletzel, Guernon, Pape), Center for Innovation for Complex Chronic Healthcare (Herrold, aronson, Kletzel, Pape), and Spinal Cord Injury/Disorder Service (Stika), Edward Hines, Jr., Veterans Affairs (VA) Hospital, Hines, IL; School of Education, Nevada State College, Henderson (Lindsey); Department of Psychology, Illinois Institute of Technology, Chicago (Ellison); Departments of Psychiatry and Behavioral Medicine (Herrold, Aaronson) and Physical Medicine and Rehabilitation (Pape), Feinberg School of Medicine, Northwestern University, Chicago; Speech-Language Pathology Program, College of Nursing and Health Sciences, Lewis University, Romeoville, IL (Guernon)
| | - Monica M Stika
- Research Service (Lindsey, Ellison, Herrold, Kletzel, Guernon, Pape), Center for Innovation for Complex Chronic Healthcare (Herrold, aronson, Kletzel, Pape), and Spinal Cord Injury/Disorder Service (Stika), Edward Hines, Jr., Veterans Affairs (VA) Hospital, Hines, IL; School of Education, Nevada State College, Henderson (Lindsey); Department of Psychology, Illinois Institute of Technology, Chicago (Ellison); Departments of Psychiatry and Behavioral Medicine (Herrold, Aaronson) and Physical Medicine and Rehabilitation (Pape), Feinberg School of Medicine, Northwestern University, Chicago; Speech-Language Pathology Program, College of Nursing and Health Sciences, Lewis University, Romeoville, IL (Guernon)
| | - Ann Guernon
- Research Service (Lindsey, Ellison, Herrold, Kletzel, Guernon, Pape), Center for Innovation for Complex Chronic Healthcare (Herrold, aronson, Kletzel, Pape), and Spinal Cord Injury/Disorder Service (Stika), Edward Hines, Jr., Veterans Affairs (VA) Hospital, Hines, IL; School of Education, Nevada State College, Henderson (Lindsey); Department of Psychology, Illinois Institute of Technology, Chicago (Ellison); Departments of Psychiatry and Behavioral Medicine (Herrold, Aaronson) and Physical Medicine and Rehabilitation (Pape), Feinberg School of Medicine, Northwestern University, Chicago; Speech-Language Pathology Program, College of Nursing and Health Sciences, Lewis University, Romeoville, IL (Guernon)
| | - Theresa Bender Pape
- Research Service (Lindsey, Ellison, Herrold, Kletzel, Guernon, Pape), Center for Innovation for Complex Chronic Healthcare (Herrold, aronson, Kletzel, Pape), and Spinal Cord Injury/Disorder Service (Stika), Edward Hines, Jr., Veterans Affairs (VA) Hospital, Hines, IL; School of Education, Nevada State College, Henderson (Lindsey); Department of Psychology, Illinois Institute of Technology, Chicago (Ellison); Departments of Psychiatry and Behavioral Medicine (Herrold, Aaronson) and Physical Medicine and Rehabilitation (Pape), Feinberg School of Medicine, Northwestern University, Chicago; Speech-Language Pathology Program, College of Nursing and Health Sciences, Lewis University, Romeoville, IL (Guernon)
| |
Collapse
|
16
|
Motanis H, Khorasani LN, Giza CC, Harris NG. Peering into the Brain through the Retrosplenial Cortex to Assess Cognitive Function of the Injured Brain. Neurotrauma Rep 2021; 2:564-580. [PMID: 34901949 PMCID: PMC8655812 DOI: 10.1089/neur.2021.0044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
The retrosplenial cortex (RSC) is a posterior cortical area that has been drawing increasing interest in recent years, with a growing number of studies studying its contribution to cognitive and sensory functions. From an anatomical perspective, it has been established that the RSC is extensively and often reciprocally connected with the hippocampus, neocortex, and many midbrain regions. Functionally, the RSC is an important hub of the default-mode network. This endowment, with vast anatomical and functional connections, positions the RSC to play an important role in episodic memory, spatial and contextual learning, sensory-cognitive activities, and multi-modal sensory information processing and integration. Additionally, RSC dysfunction has been reported in cases of cognitive decline, particularly in Alzheimer's disease and stroke. We review the literature to examine whether the RSC can act as a cortical marker of persistent cognitive dysfunction after traumatic brain injury (TBI). Because the RSC is easily accessible at the brain's surface using in vivo techniques, we argue that studying RSC network activity post-TBI can shed light into the mechanisms of less-accessible brain regions, such as the hippocampus. There is a fundamental gap in the TBI field about the microscale alterations occurring post-trauma, and by studying the RSC's neuronal activity at the cellular level we will be able to design better therapeutic tools. Understanding how neuronal activity and interactions produce normal and abnormal activity in the injured brain is crucial to understanding cognitive dysfunction. By using this approach, we expect to gain valuable insights to better understand brain disorders like TBI.
Collapse
Affiliation(s)
- Helen Motanis
- UCLA Brain Injury Research Center, Department of Neurosurgery, Geffen Medical School, UCLA Mattel Children's Hospital, University of California at Los Angeles, Los Angeles, California, USA
| | - Laila N. Khorasani
- UCLA Brain Injury Research Center, Department of Neurosurgery, Geffen Medical School, UCLA Mattel Children's Hospital, University of California at Los Angeles, Los Angeles, California, USA
| | - Christopher C. Giza
- UCLA Brain Injury Research Center, Department of Neurosurgery, Geffen Medical School, UCLA Mattel Children's Hospital, University of California at Los Angeles, Los Angeles, California, USA
- Department of Pediatrics, UCLA Mattel Children's Hospital, University of California at Los Angeles, Los Angeles, California, USA
| | - Neil G. Harris
- UCLA Brain Injury Research Center, Department of Neurosurgery, Geffen Medical School, UCLA Mattel Children's Hospital, University of California at Los Angeles, Los Angeles, California, USA
- Intellectual Development and Disabilities Research Center, UCLA Mattel Children's Hospital, University of California at Los Angeles, Los Angeles, California, USA
- *Address correspondence to: Neil G. Harris, PhD, Department of Neurosurgery, University of California at Los Angeles, Wasserman Building, 300 Stein Plaza, Room 551, Los Angeles, CA 90095, USA;
| |
Collapse
|
17
|
Multimodal characterization of Yucatan minipig behavior and physiology through maturation. Sci Rep 2021; 11:22688. [PMID: 34811385 PMCID: PMC8608884 DOI: 10.1038/s41598-021-00782-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Accepted: 10/13/2021] [Indexed: 01/11/2023] Open
Abstract
Brain injuries induced by external forces are particularly challenging to model experimentally. In recent decades, the domestic pig has been gaining popularity as a highly relevant animal model to address the pathophysiological mechanisms and the biomechanics associated with head injuries. Understanding cognitive, motor, and sensory aspects of pig behavior throughout development is crucial for evaluating cognitive and motor deficits after injury. We have developed a comprehensive battery of tests to characterize the behavior and physiological function of the Yucatan minipig throughout maturation. Behavioral testing included assessments of learning and memory, executive functions, circadian rhythms, gait analysis, and level of motor activity. We applied traditional behavioral apparatus and analysis methods, as well as state-of-the-art sensor technologies to report on motion and activity, and artificial intelligent approaches to analyze behavior. We studied pigs from 16 weeks old through sexual maturity at 35 weeks old. The results show multidimensional characterization of minipig behavior, and how it develops and changes with age. This animal model may capitulate the biomechanical consideration and phenotype of head injuries in the developing brain and can drive forward the field of understanding pathophysiological mechanisms and developing new therapies to accelerate recovery in children who have suffered head trauma.
Collapse
|
18
|
Liu J, Zheng J, Xu Y, Cao W, Wang J, Wang B, Zhao L, Zhang X, Liao W. Enriched Environment Attenuates Pyroptosis to Improve Functional Recovery After Cerebral Ischemia/Reperfusion Injury. Front Aging Neurosci 2021; 13:717644. [PMID: 34646128 PMCID: PMC8504677 DOI: 10.3389/fnagi.2021.717644] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Accepted: 09/01/2021] [Indexed: 12/16/2022] Open
Abstract
Enriched environment (EE) is a complex containing social, cognitive, and motor stimuli. Exposure to EE can promote functional recovery after ischemia/reperfusion (I/R) injury. However, the underlying mechanisms remained unclear. Pyroptosis has recently been identified and demonstrated a significant role in ischemic stroke. The purpose of this study was to explore the effect of EE on neuronal pyroptosis after cerebral I/R injury. In the current study, middle cerebral artery occlusion/reperfusion (MCAO/R) was applied to establish the cerebral I/R injury model. Behavior tests including the modified Neurological Severity Scores (mNSS) and the Morris Water Maze (MWM) were performed. The infarct volume was evaluated by Nissl staining. To evaluate the levels of pyroptosis-related proteins, the levels of GSDMD-N and nod-like receptor protein 1/3 (NLRP1/3) inflammasome-related proteins were examined. The mRNA levels of IL-1β and IL-18 were detected by Quantitative Real-Time PCR (qPCR). The secretion levels of IL-1β and IL-18 were analyzed by ELISA. Also, the expression of p65 and p-p65 were detected. The results showed that EE treatment improved functional recovery, reduced infarct volume, attenuated neuronal pyroptosis after cerebral I/R injury. EE treatment also suppressed the activities of NLRP1/NLRP3 inflammasomes. These may be affected by inhabiting the NF-κB p65 signaling pathway. Our findings suggested that neuronal pyroptosis was probably the neuroprotective mechanism that EE treatment rescued neurological deficits after I/R injury.
Collapse
Affiliation(s)
- Jingying Liu
- Department of Rehabilitation Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Jun Zheng
- Department of Rehabilitation Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Yang Xu
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Wenyue Cao
- Department of Rehabilitation Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Jinchen Wang
- Department of Anesthesiology, Zhujiang Hospital of Southern Medical University, Guangzhou, China
| | - Biru Wang
- Department of Rehabilitation Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Linyao Zhao
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xin Zhang
- Department of Rehabilitation Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Weijing Liao
- Department of Rehabilitation Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
19
|
Faillot M, Chaillet A, Palfi S, Senova S. Rodent models used in preclinical studies of deep brain stimulation to rescue memory deficits. Neurosci Biobehav Rev 2021; 130:410-432. [PMID: 34437937 DOI: 10.1016/j.neubiorev.2021.08.012] [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: 02/08/2021] [Revised: 08/10/2021] [Accepted: 08/13/2021] [Indexed: 11/28/2022]
Abstract
Deep brain stimulation paradigms might be used to treat memory disorders in patients with stroke or traumatic brain injury. However, proof of concept studies in animal models are needed before clinical translation. We propose here a comprehensive review of rodent models for Traumatic Brain Injury and Stroke. We systematically review the histological, behavioral and electrophysiological features of each model and identify those that are the most relevant for translational research.
Collapse
Affiliation(s)
- Matthieu Faillot
- Neurosurgery department, Henri Mondor University Hospital, APHP, DMU CARE, Université Paris Est Créteil, Mondor Institute for Biomedical Research, INSERM U955, Team 15, Translational Neuropsychiatry, France
| | - Antoine Chaillet
- Laboratoire des Signaux et Systèmes (L2S-UMR8506) - CentraleSupélec, Université Paris Saclay, Institut Universitaire de France, France
| | - Stéphane Palfi
- Neurosurgery department, Henri Mondor University Hospital, APHP, DMU CARE, Université Paris Est Créteil, Mondor Institute for Biomedical Research, INSERM U955, Team 15, Translational Neuropsychiatry, France
| | - Suhan Senova
- Neurosurgery department, Henri Mondor University Hospital, APHP, DMU CARE, Université Paris Est Créteil, Mondor Institute for Biomedical Research, INSERM U955, Team 15, Translational Neuropsychiatry, France.
| |
Collapse
|
20
|
Gresita A, Mihai R, Hermann DM, Amandei FS, Capitanescu B, Popa-Wagner A. Effect of environmental enrichment and isolation on behavioral and histological indices following focal ischemia in old rats. GeroScience 2021; 44:211-228. [PMID: 34382128 PMCID: PMC8811116 DOI: 10.1007/s11357-021-00432-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Accepted: 08/04/2021] [Indexed: 01/27/2024] Open
Abstract
Stroke is a disease of aging. In stroke patients, the enriched group that received stimulating physical, eating, socializing, and group activities resulted in higher activity levels including spending more time on upper limb, communal socializing, listening and iPad activities. While environmental enrichment has been shown to improve the behavioral outcome of stroke in young animals, the effect of an enriched environment on behavioral recuperation and histological markers of cellular proliferation, neuroinflammation, and neurogenesis in old subjects is not known. We used behavioral testing and immunohistochemistry to assess the effect of environment on post-stroke recovery of young and aged rats kept either in isolation or stimulating social, motor, and sensory environment (( +)Env). We provide evidence that post-stroke animals environmental enrichment ( +)Env had a significant positive effect on recovery on the rotating pole, the inclined plane, and the labyrinth test. Old age exerted a small but significant effect on lesion size, which was independent of the environment. Further, a smaller infarct volume positively correlated with better recovery of spatial learning based on positive reinforcement, working and reference memory of young, and to a lesser extent, old animals kept in ( +)Env. Histologically, isolation/impoverishment was associated with an increased number of proliferating inflammatory cells expressing ED1 cells in the peri-infarcted area of old but not young rats. Further, ( +)Env and young age were associated with an increased number of neuroepithelial cells expressing nestin/BrdU as well as beta III tubulin cells in the damaged brain area which correlated with an increased performance on the inclined plane and rotating pole. Finally, ( +)Env and an increased number of neurons expressing doublecortin/BrdU cells exerted a significant effect on performance for working memory and performance on the rotating pole in both age groups. A stimulating social, motor and sensory environment had a limited beneficial effect on behavioral recovery (working memory and rotating pole) after stroke in old rats by reducing neuroinflammation and increasing the number of neuronal precursors expressing doublecortin. Old age however, exerted a small but significant effect on lesion size, which was independent of the environment.
Collapse
Affiliation(s)
- Andrei Gresita
- Doctoral School, University of Medicine and Pharmacy, Craiova, Romania
| | - Ruscu Mihai
- Doctoral School, University of Medicine and Pharmacy, Craiova, Romania
| | - Dirk M Hermann
- Department of Neurology Chair of Vascular Neurology and Dementia, University of Medicine Essen, Essen, Germany
| | | | | | - Aurel Popa-Wagner
- Department of Neurology Chair of Vascular Neurology and Dementia, University of Medicine Essen, Essen, Germany. .,Griffith University Menzies Health Institute of Queensland, Gold Coast Campus, Southport, QLD, 4222, Australia. .,Doctoral School, University of Medicine and Pharmacy, Craiova, Romania.
| |
Collapse
|
21
|
Zhong M, Cywiak C, Metto AC, Liu X, Qian C, Pelled G. Multi-session delivery of synchronous rTMS and sensory stimulation induces long-term plasticity. Brain Stimul 2021; 14:884-894. [PMID: 34029768 DOI: 10.1016/j.brs.2021.05.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 04/17/2021] [Accepted: 05/12/2021] [Indexed: 10/21/2022] Open
Abstract
BACKGROUND Combining training or sensory stimulation with non-invasive brain stimulation has shown to improve performance in healthy subjects and improve brain function in patients after brain injury. However, the plasticity mechanisms and the optimal parameters to induce long-term and sustainable enhanced performance remain unknown. OBJECTIVE This work was designed to identify the protocols of which combining sensory stimulation with repetitive transcranial magnetic stimulation (rTMS) will facilitate the greatest changes in fMRI activation maps in the rat's primary somatosensory cortex (S1). METHODS Several protocols of combining forepaw electrical stimulation with rTMS were tested, including a single stimulation session compared to multiple, daily stimulation sessions, as well as synchronous and asynchronous delivery of both modalities. High-resolution fMRI was used to determine how pairing sensory stimulation with rTMS induced short and long-term plasticity in the rat S1. RESULTS All groups that received a single session of rTMS showed short-term increases in S1 activity, but these increases did not last three days after the session. The group that received a stimulation protocol of 10 Hz forepaw stimulation that was delivered simultaneously with 10 Hz rTMS for five consecutive days demonstrated the greatest increases in the extent of the evoked fMRI responses compared to groups that received other stimulation protocols. CONCLUSIONS Our results provide direct indication that pairing peripheral stimulation with rTMS induces long-term plasticity, and this phenomenon appears to follow a time-dependent plasticity mechanism. These results will be important to lead the design of new training and rehabilitation paradigms and training towards achieving maximal performance in healthy subjects.
Collapse
Affiliation(s)
- Ming Zhong
- Neuroengineering Division, The Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI, USA
| | - Carolina Cywiak
- Neuroengineering Division, The Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI, USA; Department of Biomedical Engineering, Michigan State University, East Lansing, MI, USA
| | - Abigael C Metto
- Neuroengineering Division, The Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI, USA; Department of Biomedical Engineering, Michigan State University, East Lansing, MI, USA
| | - Xiang Liu
- Neuroengineering Division, The Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI, USA
| | - Chunqi Qian
- Department of Radiology, Michigan State University, East Lansing, MI, USA
| | - Galit Pelled
- Neuroengineering Division, The Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI, USA; Department of Biomedical Engineering, Michigan State University, East Lansing, MI, USA; Department of Radiology, Michigan State University, East Lansing, MI, USA.
| |
Collapse
|
22
|
Cywiak C, Ashbaugh RC, Metto AC, Udpa L, Qian C, Gilad AA, Reimers M, Zhong M, Pelled G. Non-invasive neuromodulation using rTMS and the electromagnetic-perceptive gene (EPG) facilitates plasticity after nerve injury. Brain Stimul 2020; 13:1774-1783. [PMID: 33068795 DOI: 10.1016/j.brs.2020.10.006] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2020] [Revised: 10/05/2020] [Accepted: 10/12/2020] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Twenty million Americans suffer from peripheral nerve injury. These patients often develop chronic pain and sensory dysfunctions. In the past decade, neuroimaging studies showed that these changes are associated with altered cortical excitation-inhibition balance and maladaptive plasticity. We tested if neuromodulation of the deprived sensory cortex could restore the cortical balance, and whether it would be effective in alleviating sensory complications. OBJECTIVE We tested if non-invasive repetitive transcranial magnetic stimulation (rTMS) which induces neuronal excitability, and cell-specific magnetic activation via the Electromagnetic-perceptive gene (EPG) which is a novel gene that was identified and cloned from glass catfish and demonstrated to evoke neural responses when magnetically stimulated, can restore cortical excitability. METHODS A rat model of forepaw denervation was used. rTMS was delivered every other day for 30 days, starting at the acute or at the chronic post-injury phase. A minimally-invasive neuromodulation via EPG was performed every day for 30 days starting at the chronic phase. A battery of behavioral tests was performed in the days and weeks following limb denervation in EPG-treated rats, and behavioral tests, fMRI and immunochemistry were performed in rTMS-treated rats. RESULTS The results demonstrate that neuromodulation significantly improved long-term mobility, decreased anxiety and enhanced neuroplasticity. The results identify that both acute and delayed rTMS intervention facilitated rehabilitation. Moreover, the results implicate EPG as an effective cell-specific neuromodulation approach. CONCLUSION Together, these results reinforce the growing amount of evidence from human and animal studies that are establishing neuromodulation as an effective strategy to promote plasticity and rehabilitation.
Collapse
Affiliation(s)
- Carolina Cywiak
- Department of Biomedical Engineering, Michigan State University, East Lansing, MI, USA; The Institute of Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI, USA
| | - Ryan C Ashbaugh
- The Institute of Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI, USA; Department of Electrical and Computer Engineering, Michigan State University, East Lansing, MI, USA
| | - Abigael C Metto
- Department of Biomedical Engineering, Michigan State University, East Lansing, MI, USA; The Institute of Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI, USA
| | - Lalita Udpa
- Department of Electrical and Computer Engineering, Michigan State University, East Lansing, MI, USA
| | - Chunqi Qian
- Department of Radiology, Michigan State University, East Lansing, MI, USA
| | - Assaf A Gilad
- Department of Biomedical Engineering, Michigan State University, East Lansing, MI, USA; The Institute of Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI, USA; Department of Radiology, Michigan State University, East Lansing, MI, USA
| | - Mark Reimers
- The Institute of Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI, USA; Department of Physiology and Neuroscience Program, Michigan State University, East Lansing, MI, USA
| | - Ming Zhong
- The Institute of Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI, USA
| | - Galit Pelled
- Department of Biomedical Engineering, Michigan State University, East Lansing, MI, USA; The Institute of Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI, USA; Department of Radiology, Michigan State University, East Lansing, MI, USA.
| |
Collapse
|
23
|
Walter J, Kovalenko O, Younsi A, Grutza M, Unterberg A, Zweckberger K. The CatWalk XT® is a valid tool for objective assessment of motor function in the acute phase after controlled cortical impact in mice. Behav Brain Res 2020; 392:112680. [DOI: 10.1016/j.bbr.2020.112680] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2019] [Revised: 04/15/2020] [Accepted: 04/27/2020] [Indexed: 01/01/2023]
|
24
|
Nardone R, Sebastianelli L, Versace V, Brigo F, Golaszewski S, Manganotti P, Saltuari L, Trinka E. Repetitive transcranial magnetic stimulation in traumatic brain injury: Evidence from animal and human studies. Brain Res Bull 2020; 159:44-52. [PMID: 32251693 DOI: 10.1016/j.brainresbull.2020.03.016] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2019] [Revised: 03/16/2020] [Accepted: 03/23/2020] [Indexed: 12/19/2022]
Abstract
We provide here the first systematic review on the studies dealing with repetitive transcranial magnetic stimulation (rTMS) for traumatic brain injury (TBI) in animals and humans. Several experimental studies in animal models have explored with promising results the use of rTMS to enhance neuroprotection and recovery after TBI. However, there are surprisingly few studies that have obtained substantial evidence regarding effects of rTMS in humans with TBI, many of them are case reports investigating the heterogeneous conditions linked to TBI. The most studies have investigated the effects of rTMS in subjects with post-traumatic depression and variable effects have been observed. rTMS has been proposed as an experimental approach for the treatment of disorders of consciousness (DOC), but in subjects with TBI therapeutic effects on DOC have also been variously documented. Beneficial effects have been reported in subjects with cognitive/emotional disturbances and auditory dysfunction (tinnitus and hallucinations), although the results are somewhat conflicting. rTMS applied over the left prefrontal cortex may relieve, at least transiently, post-traumatic headache. Isolated rTMS studies have been performed in TBI patients with motor impairment, chronic dizziness or pain. Especially whether provided in combination, rTMS and neurorehabilitation may be synergistic in the potential to translate experimental findings in the clinical practice. In order to reach definitive conclusions, well-designed randomized controlled studies with larger patient samples, improved design and optimized rTMS setup, are warranted to verify and corroborate the initial promising findings.
Collapse
Affiliation(s)
- Raffaele Nardone
- Department of Neurology, Franz Tappeiner Hospital, Merano, Italy; Department of Neurology, Christian Doppler Klinik, Paracelsus Medical University, Salzburg, Austria; Spinal Cord Injury and Tissue Regeneration Center, Salzburg, Austria; Karl Landsteiner Institut für Neurorehabilitation und Raumfahrtneurologie, Salzburg, Austria.
| | - Luca Sebastianelli
- Department of Neurorehabilitation, Hospital of Vipiteno, Vipiteno, Italy; Research Unit for Neurorehabilitation South Tyrol, Bolzano, Italy
| | - Viviana Versace
- Department of Neurorehabilitation, Hospital of Vipiteno, Vipiteno, Italy; Research Unit for Neurorehabilitation South Tyrol, Bolzano, Italy
| | - Francesco Brigo
- Department of Neurology, Franz Tappeiner Hospital, Merano, Italy; Department of Neuroscience, Biomedicine and Movement Science, University of Verona, Italy
| | - Stefan Golaszewski
- Department of Neurology, Christian Doppler Klinik, Paracelsus Medical University, Salzburg, Austria; Karl Landsteiner Institut für Neurorehabilitation und Raumfahrtneurologie, Salzburg, Austria
| | | | - Leopold Saltuari
- Department of Neurorehabilitation, Hospital of Vipiteno, Vipiteno, Italy; Research Unit for Neurorehabilitation South Tyrol, Bolzano, Italy
| | - Eugen Trinka
- Department of Neurology, Christian Doppler Klinik, Paracelsus Medical University, Salzburg, Austria; Centre for Cognitive Neurosciences Salzburg, Salzburg, Austria; University for Medical Informatics and Health Technology, UMIT, Hall in Tirol, Austria
| |
Collapse
|
25
|
Abstract
Developments of new strategies to restore vision and improving on current strategies by harnessing new advancements in material and electrical sciences, and biological and genetic-based technologies are of upmost health priorities around the world. Federal and private entities are spending billions of dollars on visual prosthetics technologies. This review describes the most current and state-of-the-art bioengineering technologies to restore vision. This includes a thorough description of traditional electrode-based visual prosthetics that have improved substantially since early prototypes. Recent advances in molecular and synthetic biology have transformed vision-assisted technologies; For example, optogenetic technologies that introduce light-responsive proteins offer excellent resolution but cortical applications are restricted by fiber implantation and tissue damage. Other stimulation modalities, such as magnetic fields, have been explored to achieve non-invasive neuromodulation. Miniature magnetic coils are currently being developed to activate select groups of neurons. Magnetically-responsive nanoparticles or exogenous proteins can significantly enhance the coupling between external electromagnetic devices and any neurons affiliated with these modifications. The need to minimize cytotoxic effects for nanoparticle-based therapies will likely restrict the number of usable materials. Nevertheless, advances in identifying and utilizing proteins that respond to magnetic fields may lead to non-invasive, cell-specific stimulation and may overcome many of the limitations that currently exist with other methods. Finally, sensory substitution systems also serve as viable visual prostheses by converting visual input to auditory and somatosensory stimuli. This review also discusses major challenges in the field and offers bioengineering strategies to overcome those.
Collapse
Affiliation(s)
- Alexander Farnum
- Department of Biomedical Engineering, College of Engineering, Michigan State University, East Lansing, MI, United States
- Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI, United States
| | - Galit Pelled
- Department of Biomedical Engineering, College of Engineering, Michigan State University, East Lansing, MI, United States
- Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI, United States
- Department of Radiology, College of Human Medicine, Michigan State University, East Lansing, MI, United States
| |
Collapse
|
26
|
Zhao CG, Qin J, Sun W, Ju F, Zhao YL, Wang R, Sun XL, Mou X, Yuan H. rTMS Regulates the Balance Between Proliferation and Apoptosis of Spinal Cord Derived Neural Stem/Progenitor Cells. Front Cell Neurosci 2020; 13:584. [PMID: 32116552 PMCID: PMC7025559 DOI: 10.3389/fncel.2019.00584] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Accepted: 12/20/2019] [Indexed: 01/31/2023] Open
Abstract
Repetitive transcranial magnetic stimulation (rTMS) is a noninvasive technique that uses electromagnetic fields to stimulate the brain. rTMS can restore an impaired central nervous system and promote proliferation of neural stem/progenitor cells (NSPCs), but optimal stimulus parameters and mechanisms underlying these effects remain elusive. The purpose of this study is to investigate the effect of different rTMS stimulus parameters on proliferation and apoptosis of spinal cord-derived NSPCs, the expression of brain-derived neurotrophic factor (BDNF) after rTMS, and the potentially underlying pathways. NSPCs were isolated from mice spinal cord and stimulated by different frequencies (1/10/20 Hz), intensities (0.87/1.24/1.58 T), and number of pulses (400/800/1,500/3,000) once a day for five consecutive days. NSPC proliferation was analyzed by measuring the neurosphere diameter and Brdu staining, apoptosis was detected by cell death enzyme-linked immunosorbent assay (ELISA) and flow cytometry, and NSPC viability was assessed by cell counting kit-8 assay. We found that specific parameters of frequency (1/10/20 Hz), intensity (1.24/1.58 T), and number of pulses (800/1,500/3,000) promote proliferation and apoptosis (p < 0.05 for all), but 20 Hz, 1.58 T, and 1,500 pulses achieved the optimal response for the NSPC viability. In addition, rTMS significantly promoted the expression of BDNF at the mRNA and protein level, while also increasing Akt phosphorylation (Thr308 and Ser473; p < 0.05). Overall, we identified the most appropriate rTMS parameters for further studies on NSPCs in vitro and in vivo. Furthermore, the effect of magnetic stimulation on NSPC proliferation might be correlated to BDNF/Akt signaling pathway.
Collapse
Affiliation(s)
- Chen-Guang Zhao
- Department of Rehabilitation Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Jie Qin
- Department of Orthopedics, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Wei Sun
- Department of Rehabilitation Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Fen Ju
- Department of Rehabilitation Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Yong-Lin Zhao
- Department of Oncology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Rui Wang
- Department of Orthopedics, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Xiao-Long Sun
- Department of Rehabilitation Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Xiang Mou
- Department of Rehabilitation Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Hua Yuan
- Department of Rehabilitation Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| |
Collapse
|
27
|
Meyers EC, Kasliwal N, Solorzano BR, Lai E, Bendale G, Berry A, Ganzer PD, Romero-Ortega M, Rennaker RL, Kilgard MP, Hays SA. Enhancing plasticity in central networks improves motor and sensory recovery after nerve damage. Nat Commun 2019; 10:5782. [PMID: 31857587 PMCID: PMC6923364 DOI: 10.1038/s41467-019-13695-0] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Accepted: 11/08/2019] [Indexed: 12/11/2022] Open
Abstract
Nerve damage can cause chronic, debilitating problems including loss of motor control and paresthesia, and generates maladaptive neuroplasticity as central networks attempt to compensate for the loss of peripheral connectivity. However, it remains unclear if this is a critical feature responsible for the expression of symptoms. Here, we use brief bursts of closed-loop vagus nerve stimulation (CL-VNS) delivered during rehabilitation to reverse the aberrant central plasticity resulting from forelimb nerve transection. CL-VNS therapy drives extensive synaptic reorganization in central networks paralleled by improved sensorimotor recovery without any observable changes in the nerve or muscle. Depleting cortical acetylcholine blocks the plasticity-enhancing effects of CL-VNS and consequently eliminates recovery, indicating a critical role for brain circuits in recovery. These findings demonstrate that manipulations to enhance central plasticity can improve sensorimotor recovery and define CL-VNS as a readily translatable therapy to restore function after nerve damage. Peripheral nerve damage generates maladaptive neuroplasticity as central networks attempt to compensate for the loss of peripheral connectivity. Here, the authors reverse the aberrant plasticity via vagus nerve stimulation to elicit synaptic reorganization and to improve sensorimotor recovery.
Collapse
Affiliation(s)
- Eric C Meyers
- Texas Biomedical Device Center, The University of Texas at Dallas, 800 West Campbell Road, Richardson, TX, 75080-3021, USA.
| | - Nimit Kasliwal
- Texas Biomedical Device Center, The University of Texas at Dallas, 800 West Campbell Road, Richardson, TX, 75080-3021, USA
| | - Bleyda R Solorzano
- Texas Biomedical Device Center, The University of Texas at Dallas, 800 West Campbell Road, Richardson, TX, 75080-3021, USA
| | - Elaine Lai
- School of Behavioral and Brain Sciences, The University of Texas at Dallas, 800 West Campbell Road, Richardson, TX, 75080-3021, USA
| | - Geetanjali Bendale
- Department of Bioengineering, Erik Jonsson School of Engineering and Computer Science, The University of Texas at Dallas, 800 West Campbell Road, Richardson, TX, 75080-3021, USA
| | - Abigail Berry
- Texas Biomedical Device Center, The University of Texas at Dallas, 800 West Campbell Road, Richardson, TX, 75080-3021, USA
| | - Patrick D Ganzer
- Texas Biomedical Device Center, The University of Texas at Dallas, 800 West Campbell Road, Richardson, TX, 75080-3021, USA
| | - Mario Romero-Ortega
- Texas Biomedical Device Center, The University of Texas at Dallas, 800 West Campbell Road, Richardson, TX, 75080-3021, USA.,School of Behavioral and Brain Sciences, The University of Texas at Dallas, 800 West Campbell Road, Richardson, TX, 75080-3021, USA.,Department of Bioengineering, Erik Jonsson School of Engineering and Computer Science, The University of Texas at Dallas, 800 West Campbell Road, Richardson, TX, 75080-3021, USA
| | - Robert L Rennaker
- Texas Biomedical Device Center, The University of Texas at Dallas, 800 West Campbell Road, Richardson, TX, 75080-3021, USA.,School of Behavioral and Brain Sciences, The University of Texas at Dallas, 800 West Campbell Road, Richardson, TX, 75080-3021, USA.,Department of Bioengineering, Erik Jonsson School of Engineering and Computer Science, The University of Texas at Dallas, 800 West Campbell Road, Richardson, TX, 75080-3021, USA
| | - Michael P Kilgard
- Texas Biomedical Device Center, The University of Texas at Dallas, 800 West Campbell Road, Richardson, TX, 75080-3021, USA.,School of Behavioral and Brain Sciences, The University of Texas at Dallas, 800 West Campbell Road, Richardson, TX, 75080-3021, USA.,Department of Bioengineering, Erik Jonsson School of Engineering and Computer Science, The University of Texas at Dallas, 800 West Campbell Road, Richardson, TX, 75080-3021, USA
| | - Seth A Hays
- Texas Biomedical Device Center, The University of Texas at Dallas, 800 West Campbell Road, Richardson, TX, 75080-3021, USA.,School of Behavioral and Brain Sciences, The University of Texas at Dallas, 800 West Campbell Road, Richardson, TX, 75080-3021, USA.,Department of Bioengineering, Erik Jonsson School of Engineering and Computer Science, The University of Texas at Dallas, 800 West Campbell Road, Richardson, TX, 75080-3021, USA
| |
Collapse
|
28
|
Frequency-specific effects of low-intensity rTMS can persist for up to 2 weeks post-stimulation: A longitudinal rs-fMRI/MRS study in rats. Brain Stimul 2019; 12:1526-1536. [PMID: 31296402 DOI: 10.1016/j.brs.2019.06.028] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Revised: 06/23/2019] [Accepted: 06/26/2019] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Evidence suggests that repetitive transcranial magnetic stimulation (rTMS), a non-invasive neuromodulation technique, alters resting brain activity. Despite anecdotal evidence that rTMS effects wear off, there are no reports of longitudinal studies, even in humans, mapping the therapeutic duration of rTMS effects. OBJECTIVE Here, we investigated the longitudinal effects of repeated low-intensity rTMS (LI-rTMS) on healthy rodent resting-state networks (RSNs) using resting-state functional MRI (rs-fMRI) and on sensorimotor cortical neurometabolite levels using proton magnetic resonance spectroscopy (MRS). METHODS Sprague-Dawley rats received 10 min LI-rTMS daily for 15 days (10 Hz or 1 Hz stimulation, n = 9 per group). MRI data were acquired at baseline, after seven days and after 14 days of daily stimulation and at two more timepoints up to three weeks post-cessation of daily stimulation. RESULTS 10 Hz stimulation increased RSN connectivity and GABA, glutamine, and glutamate levels. 1 Hz stimulation had opposite but subtler effects, resulting in decreased RSN connectivity and glutamine levels. The induced changes decreased to baseline levels within seven days following stimulation cessation in the 10 Hz group but were sustained for at least 14 days in the 1 Hz group. CONCLUSION Overall, our study provides evidence of long-term frequency-specific effects of LI-rTMS. Additionally, the transient connectivity changes following 10 Hz stimulation suggest that current treatment protocols involving this frequency may require ongoing "top-up" stimulation sessions to maintain therapeutic effects.
Collapse
|
29
|
Krishnan VS, Shin SS, Belegu V, Celnik P, Reimers M, Smith KR, Pelled G. Multimodal Evaluation of TMS - Induced Somatosensory Plasticity and Behavioral Recovery in Rats With Contusion Spinal Cord Injury. Front Neurosci 2019; 13:387. [PMID: 31068784 PMCID: PMC6491761 DOI: 10.3389/fnins.2019.00387] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Accepted: 04/04/2019] [Indexed: 12/16/2022] Open
Abstract
Introduction: Spinal cord injury (SCI) causes partial or complete damage to sensory and motor pathways and induces immediate changes in cortical function. Current rehabilitative strategies do not address this early alteration, therefore impacting the degree of neuroplasticity and subsequent recovery. The following study aims to test if a non-invasive brain stimulation technique such as repetitive transcranial magnetic stimulation (rTMS) is effective in promoting plasticity and rehabilitation, and can be used as an early intervention strategy in a rat model of SCI. Methods: A contusion SCI was induced at segment T9 in adult rats. An rTMS coil was positioned over the brain to deliver high frequency stimulation. Behavior, motor and sensory functions were tested in three groups: SCI rats that received high-frequency (20 Hz) rTMS within 10 min post-injury (acute-TMS; n = 7); SCI rats that received TMS starting 2 weeks post-injury (chronic-TMS; n = 5), and SCI rats that received sham TMS (no-TMS, n = 5). Locomotion was evaluated by the Basso, Beattie, and Bresnahan (BBB) and gridwalk tests. Motor evoked potentials (MEP) were recorded from the forepaw across all groups to measure integrity of motor pathways. Functional MRI (fMRI) responses to contralateral tactile hindlimb stimulation were measured in an 11.7T horizontal bore small-animal scanner. Results: The acute-TMS group demonstrated the fastest improvements in locomotor performance in both the BBB and gridwalk tests compared to chronic and no-TMS groups. MEP responses from forepaw showed significantly greater difference in the inter-peak latency between acute-TMS and no-TMS groups, suggesting increases in motor function. Finally, the acute-TMS group showed increased fMRI-evoked responses to hindlimb stimulation over the right and left hindlimb (LHL) primary somatosensory representations (S1), respectively; the chronic-TMS group showed moderate sensory responses in comparison, and the no-TMS group exhibited the lowest sensory responses to both hindlimbs. Conclusion: The results suggest that rTMS therapy beginning in the acute phase after SCI promotes neuroplasticity and is an effective rehabilitative approach in a rat model of SCI.
Collapse
Affiliation(s)
- Vijai S Krishnan
- Department of Biomedical Engineering, Michigan State University, East Lansing, MI, United States.,The Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI, United States.,F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, MD, United States.,Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Samuel S Shin
- F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, MD, United States
| | - Visar Belegu
- Department of Neurology and Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, United States.,International Center for Spinal Cord Injury, Kennedy Krieger Institute, Baltimore, MD, United States
| | - Pablo Celnik
- Department of Physical Medicine and Rehabilitation, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Mark Reimers
- Department of Biomedical Engineering, Michigan State University, East Lansing, MI, United States.,The Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI, United States
| | - Kylie R Smith
- Department of Biomedical Engineering, Michigan State University, East Lansing, MI, United States.,The Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI, United States
| | - Galit Pelled
- Department of Biomedical Engineering, Michigan State University, East Lansing, MI, United States.,The Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI, United States.,F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, MD, United States.,Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, United States.,Department of Radiology, Michigan State University, East Lansing, MI, United States
| |
Collapse
|
30
|
Wang Y, Fang K, He S, Fan Y, Yu J, Zhang X. Effects of repetitive magnetic stimulation on the growth of primarily cultured hippocampus neurons in vitro and their expression of iron-containing enzymes. Neuropsychiatr Dis Treat 2019; 15:927-934. [PMID: 31114204 PMCID: PMC6489628 DOI: 10.2147/ndt.s199328] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/24/2018] [Accepted: 03/05/2019] [Indexed: 12/22/2022] Open
Abstract
Background: The mechanism of action of repetitive transcranial magnetic stimulation (rTMS) involves the generation of neuronal and action potentials utilizing induced currents in time-varying magnetic fields. However, the long-lasting and effective biological impact of magnetic stimulation does not appear to be completely explained by the transient magnetic field pulses. In this context, we hypothesized magnetic stimulation may affect the expression of iron-containing enzymes in neurons, mediating the long-lasting biological effects associated with this stimulus. Methods: Primarily cultured hippocampus neurons from SD rats were used as the cell model in this study. These were randomly divided into control, sham, and magnetic stimulation groups to probe into the effect of the magnetic field directly. The latter group received 40%, 60%, and 100% maximal stimulator output Tesla (1.68, 2.52, and 4.2 T) with low-frequency rTMS (1 Hz). The expression of iron-containing enzymes (catalase and aconitase) and non-ferrous enzymes (protein kinase A) was measured with Western blotting and ELISA. Results: The survival rates of neurons in the 40%T and 60%T groups were significantly increased in comparison to the controls (P<0.05), while those in the 100%T group showed cell damage, with slightly disturbed neurite connections and decreased survival rate. Furthermore, catalase and aconitase expression was higher in all of the stimulated groups in comparison to controls (P<0.05). On the other hand, the expression of the iron-containing enzymes decreased in the 100%T group in comparison with the 40%T and 60%T groups (P<0.05). Meanwhile, the expression of protein kinase A was not significantly increased in the groups which underwent magnetic stimulation. Conclusion: rTMS may increase the expression of ferrous enzymes but does not have a strong effect on non-ferrous enzymes. Excessive intensity of magnetic stimulation may reduce neuronal survival rate and affect the expression of iron-containing enzymes. The mechanism underlying the lasting effect of rTMS may be related to the increase of ferriferous expression induced by magnetic stimulation, with a clear correlation with stimulation intensity.
Collapse
Affiliation(s)
- Yirong Wang
- Department of Neurology, Affiliated Hospital of North Sichuan Medical College, Sichuan, People's Republic of China
| | - Kewei Fang
- Department of Radiology, Affiliated Hospital of North Sichuan Medical College, Sichuan, People's Republic of China
| | - Shijia He
- Department of Neurology, Affiliated Hospital of North Sichuan Medical College, Sichuan, People's Republic of China
| | - Yang Fan
- Department of Neurology, Affiliated Hospital of North Sichuan Medical College, Sichuan, People's Republic of China
| | - Juming Yu
- Department of Neurology, Affiliated Hospital of North Sichuan Medical College, Sichuan, People's Republic of China
| | - Xiaodong Zhang
- Department of Neurology, Affiliated Hospital of North Sichuan Medical College, Sichuan, People's Republic of China
| |
Collapse
|