1
|
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
|
2
|
Kim HM, Jo HS, Kim EJ, Na JM, Park HK, Han JY, Kim KH, Choi I, Song MK. The Effect of Repetitive Transcranial Magnetic Stimulation on Cognition in Diffuse Axonal Injury in a Rat Model. Neurol Int 2024; 16:689-700. [PMID: 39051213 PMCID: PMC11270180 DOI: 10.3390/neurolint16040052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 06/14/2024] [Accepted: 06/17/2024] [Indexed: 07/27/2024] Open
Abstract
Diffuse axonal injury (DAI) following sudden acceleration and deceleration can lead to cognitive function decline. Various treatments have been proposed. Repetitive transcranial magnetic stimulation (rTMS), a non-invasive stimulation technique, is a potential treatment for enhancing neuroplasticity in cases of brain injury. The therapeutic efficacy of rTMS on cognitive function remains unconfirmed. This study investigated the effects of rTMS and the underlying molecular biomechanisms using a rat model of DAI. Sprague-Dawley rats (n = 18) were randomly divided into two groups: one receiving rTMS after DAI and the other without brain stimulation. All rats were subjected to sudden acceleration and deceleration using a DAI modeling machine to induce damage. MRI was performed to confirm the DAI lesion. The experimental group received rTMS at a frequency of 1 Hz over the frontal cortex for 10 min daily for five days. To assess spatial memory, we conducted the Morris water maze (MWM) test one day post-brain damage and one day after the five-day intervention. A video tracking system recorded the escape latency. After post-MWM tests, all rats were euthanized, and their brain tissues, particularly from the hippocampus, were collected for immunohistochemistry and western blot analyses. The escape latency showed no difference on the MWM test after DAI, but a significant difference was observed after rTMS between the two groups. Immunohistochemistry and western blot analyses indicated increased expression of BDNF, VEGF, and MAP2 in the hippocampal brain tissue of the DAI-T group. In conclusion, rTMS improved cognitive function in the DAI rat model. The increased expression of BDNF, VEGF, and MAP2 in the DAI-T group supports the potential use of rTMS in treating cognitive impairments associated with DAI.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Insung Choi
- Department of Physical & Rehabilitation Medicine, Chonnam National University Medical School & Hospital, Gwangju 61469, Republic of Korea; (H.-M.K.); (H.-S.J.); (E.-J.K.); (J.-M.N.); (H.-K.P.); white-- (J.-Y.H.); (K.-H.K.)
| | - Min-Keun Song
- Department of Physical & Rehabilitation Medicine, Chonnam National University Medical School & Hospital, Gwangju 61469, Republic of Korea; (H.-M.K.); (H.-S.J.); (E.-J.K.); (J.-M.N.); (H.-K.P.); white-- (J.-Y.H.); (K.-H.K.)
| |
Collapse
|
3
|
McNerney MW, Gurkoff GG, Beard C, Berryhill ME. The Rehabilitation Potential of Neurostimulation for Mild Traumatic Brain Injury in Animal and Human Studies. Brain Sci 2023; 13:1402. [PMID: 37891771 PMCID: PMC10605899 DOI: 10.3390/brainsci13101402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 09/25/2023] [Accepted: 09/28/2023] [Indexed: 10/29/2023] Open
Abstract
Neurostimulation carries high therapeutic potential, accompanied by an excellent safety profile. In this review, we argue that an arena in which these tools could provide breakthrough benefits is traumatic brain injury (TBI). TBI is a major health problem worldwide, with the majority of cases identified as mild TBI (mTBI). MTBI is of concern because it is a modifiable risk factor for dementia. A major challenge in studying mTBI is its inherent heterogeneity across a large feature space (e.g., etiology, age of injury, sex, treatment, initial health status, etc.). Parallel lines of research in human and rodent mTBI can be collated to take advantage of the full suite of neuroscience tools, from neuroimaging (electroencephalography: EEG; functional magnetic resonance imaging: fMRI; diffusion tensor imaging: DTI) to biochemical assays. Despite these attractive components and the need for effective treatments, there are at least two major challenges to implementation. First, there is insufficient understanding of how neurostimulation alters neural mechanisms. Second, there is insufficient understanding of how mTBI alters neural function. The goal of this review is to assemble interrelated but disparate areas of research to identify important gaps in knowledge impeding the implementation of neurostimulation.
Collapse
Affiliation(s)
- M. Windy McNerney
- Mental Illness Research Education and Clinical Center (MIRECC), Veterans Affairs Palo Alto Health Care System, Palo Alto, CA 94304, USA; (M.W.M.); (C.B.)
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Gene G. Gurkoff
- Department of Neurological Surgery, and Center for Neuroscience, University of California, Davis, Sacramento, CA 95817, USA;
- Department of Veterans Affairs, VA Northern California Health Care System, Martinez, CA 94553, USA
| | - Charlotte Beard
- Mental Illness Research Education and Clinical Center (MIRECC), Veterans Affairs Palo Alto Health Care System, Palo Alto, CA 94304, USA; (M.W.M.); (C.B.)
- Program in Neuroscience and Behavioral Biology, Emory University, Atlanta, GA 30322, USA
| | - Marian E. Berryhill
- Programs in Cognitive and Brain Sciences, and Integrative Neuroscience, Department of Psychology, University of Nevada, Reno, NV 89557, USA
| |
Collapse
|
4
|
Makale MT, Nybo C, Keifer J, Blum K, Dennen CA, Baron D, Sunder K, Elman I, Makale MR, Thanos PK, Murphy KT. Preliminary Observations of Personalized Repetitive Magnetic Stimulation (PrTMS) Guided by EEG Spectra for Concussion. Brain Sci 2023; 13:1179. [PMID: 37626535 PMCID: PMC10452199 DOI: 10.3390/brainsci13081179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 08/02/2023] [Accepted: 08/08/2023] [Indexed: 08/27/2023] Open
Abstract
There are no FDA-approved treatments for the chronic sequelae of concussion. Repetitive magnetic transcranial stimulation (rTMS) has been explored as a therapy but outcomes have been inconsistent. To address this we developed a personalized rTMS (PrTMS) protocol involving continual rTMS stimulus frequency adjustment and progressive activation of multiple cortical sites, guided by spectral electroencephalogram (EEG)-based analyses and psychological questionnaires. We acquired pilot clinical data for 185 symptomatic brain concussion patients who underwent the PrTMS protocol over an approximate 6 week period. The PrTMS protocol used a proprietary EEG spectral frequency algorithm to define an initial stimulation frequency based on an anteriorly graded projection of the measured occipital alpha center peak, which was then used to interpolate and adjust regional stimulation frequency according to weekly EEG spectral acquisitions. PrTMS improved concussion indices and normalized the cortical alpha band center frequency and peak EEG amplitude. This potentially reflected changed neurotransmitter, cognitive, and perceptual status. PrTMS may be a promising treatment choice for patients with persistent concussion symptoms. This clinical observational study was limited in that there was no control group and a number of variables were not recorded, such as time since injury and levels of depression. While the present observations are indeed preliminary and cursory, they may suggest further prospective research on PrTMS in concussion, and exploration of the spectral EEG as a concussion biomarker, with the ultimate goals of confirmation and determining optimal PrTMS treatment parameters.
Collapse
Affiliation(s)
- Milan T. Makale
- Department of Radiation Medicine and Applied Sciences, University of California San Diego, La Jolla, CA 92093, USA
| | - Chad Nybo
- CrossTx Inc., Bozeman, MT 59715, USA
| | | | - Kenneth Blum
- Department of Clinical Psychology and Addiction, Institute of Psychology, Faculty of Education and Psychology, Eötvös Loránd University, 1075 Budapest, Hungary
- Department of Psychiatry, Wright University, Boonshoft School of Medicine, Dayton, OH 45324, USA
- Department of Molecular Biology and Adelson School of Medicine, Ariel University, Ariel 40700, Israel
- Division of Addiction Research & Education, Center for Sports, Exercise & Global Mental Health, Western University Health Sciences, Pomona, CA 91766, USA
| | - Catherine A. Dennen
- Department of Family Medicine, Jefferson Health NE, Philadelphia, PA 19107, USA
| | - David Baron
- Division of Addiction Research & Education, Center for Sports, Exercise & Global Mental Health, Western University Health Sciences, Pomona, CA 91766, USA
| | - Keerthy Sunder
- School of Medicine, University of California Riverside, Riverside, CA 92521, USA
| | - Igor Elman
- Cambridge Health Alliance, Harvard Medical School, Cambridge, MA 02143, USA
| | - Miles R. Makale
- Department of Psychology, University of California San Diego, La Jolla, CA 92093, USA
| | - Panayotis K. Thanos
- Behavioral Neuropharmacology and Neuroimaging Laboratory on Addictions, Clinical Research Institute on Addictions, Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biosciences, State University of New York at Buffalo, Buffalo, NY 14203, USA
- Department of Psychology, State University of New York at Buffalo, Buffalo, NY 14203, USA
| | | |
Collapse
|
5
|
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: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [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
|
6
|
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: 12] [Impact Index Per Article: 6.0] [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
|
7
|
Diaz MJ, Root KT, Beneke A, Penev Y, Lucke-Wold B. Neurostimulation for Traumatic Brain Injury: Emerging Innovation. OBM NEUROBIOLOGY 2023; 07:1-17. [PMID: 36938307 PMCID: PMC10019379 DOI: 10.21926/obm.neurobiol.2301161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/08/2023]
Abstract
Traumatic brain injury (TBI) is a significant source of brain deficit and death among neurosurgical patients, with limited prospects for functional recovery in the cases of moderate-to-severe injury. Until now, the relevant body of literature on TBI intervention has focused on first-line, invasive treatment options (namely craniectomy and hematoma evacuation) with underwhelming focus on non-invasive therapies following surgical stabilization. Recent advances in our understanding of the impaired brain have encouraged deeper investigation of neurostimulation strategies, owed largely to its demonstrated livening of damaged neural circuitry and capacity to stabilize erratic network activity. The objective of the present study is to provide a scoping review of new knowledge in neurostimulation published in the PubMed, Scopus, and Google Scholar databases from inception to November 2022. We critically assess and appraise the available data on primary neurostimulation delivery techniques, with marked emphasis on restorative opportunities for accessory neurostimulation in the interdisciplinary care of moderate-to-severe TBI (msTBI) patients. These data identify two primary future directions: 1) to relate obtained gain-of-function outcomes to hemodynamic and histological changes and 2) to develop a clearer understanding of neurostimulation efficacy, when combined with pharmacologic interventions or other modulatory techniques, for complex brain insult.
Collapse
|
8
|
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
|
9
|
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
|
10
|
Peng Y, Zhao Y, Huang Y, Liu X, Zhang H, Zhao Z, Cheng Y, Liu L. Neuroprotective effects of low-intensity transcranial ultrasound stimulation combined with Baicalin intervention on traumatic brain injury in animals. Brain Res Bull 2021; 175:246-253. [PMID: 34343642 DOI: 10.1016/j.brainresbull.2021.07.028] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2021] [Revised: 06/29/2021] [Accepted: 07/29/2021] [Indexed: 10/20/2022]
Abstract
Low-intensity transcranial ultrasound stimulation (LITUS) can improve the inflammatory reaction after traumatic brain injury (TBI), and Baicalin also has a good protective effect on TBI. The purpose of this study was to observe the neuroprotective effect of LITUS combined with Baicalin intervention in the TBI rats. Sprague Dawley (SD) rats were randomly divided into 5 groups (n = 15) which were Sham control group, TBI group, LITUS group, Baicalin group, LITUS combined with Baicalin group (LB group). The rats were scanned with 3.0 T magnetic resonance imager, and the apparent diffusion coefficient (ADC) and the fractional anisotropy (FA) of the brain injury cortical area were determined at 3 h, 1, 3, 7 and 10 d after TBI. The ADC value, FA value, neurological function score and Nissl staining were used to assess the level of brain damage of rats. The results showed that on the 10th day after TBI, the ADC values of the TBI group, the LITUS group and the Baicalin group were remarkable greater than that of the L-B group (all adjusted P < 0.05), FA values were remarkable smaller than that of the L-B group (all adjusted P < 0.05), neurological function scores were remarkable greater than that of the L-B group (all adjusted P < 0.05), and Nissl body loss rates were remarkable greater than that of the L-B group (all adjusted P < 0.001). This study indicated that compared with the LITUS group and the Baicalin group, the L-B group can more effectively reduce level of brain damage after TBI, and the method of LITUS combined with Baicalin intervention was a more effective neuroprotection for brain injury.
Collapse
Affiliation(s)
- Yong Peng
- Institute of Electrical Engineering, Yanshan University, China; Key Laboratory of Intelligent Rehabilitation and Neuromodulation of Hebei Province, Yanshan University, China.
| | - Yang Zhao
- Institute of Electrical Engineering, Yanshan University, China
| | - Yameng Huang
- Institute of Electrical Engineering, Yanshan University, China
| | - Xiaoyue Liu
- Institute of Electrical Engineering, Yanshan University, China
| | - Hui Zhang
- Institute of Electrical Engineering, Yanshan University, China
| | - Zheng Zhao
- Institute of Electrical Engineering, Yanshan University, China
| | - Yawei Cheng
- Institute of Electrical Engineering, Yanshan University, China
| | | |
Collapse
|
11
|
Deng Y, Guo F, Han X, Huang X. Repetitive transcranial magnetic stimulation increases neurological function and endogenous neural stem cell migration via the SDF-1α/CXCR4 axis after cerebral infarction in rats. Exp Ther Med 2021; 22:1037. [PMID: 34373723 PMCID: PMC8343462 DOI: 10.3892/etm.2021.10469] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Accepted: 06/09/2021] [Indexed: 12/21/2022] Open
Abstract
Neural stem cell (NSC) migration is closely associated with brain development and is reportedly involved during recovery from ischaemic stroke. Chemokine signalling mediated by stromal cell-derived factor 1α (SDF-1α) and its receptor CXC chemokine receptor 4 (CXCR4) has been previously documented to guide the migration of NSCs. Although repetitive transcranial magnetic stimulation (rTMS) can increase neurological function in a rat stroke model, its effects on the migration of NSCs and associated underlying mechanism remain unclear. Therefore, the present study investigated the effects of rTMS on ischaemic stroke following middle cerebral artery occlusion (MCAO). All rats underwent rTMS treatment 24 h after MCAO. Neurological function, using modified Neurological Severity Scores and grip strength test and NSC migration, which were measured using immunofluorescence staining, were analysed at 7 and 14 days after MCAO, before the protein expression levels of the SDF-1α/CXCR4 axis was evaluated using western blot analysis. AMD3100, a CXCR4 inhibitor, was used to assess the effects of SDF-1α/CXCR4 signalling. In addition, neuronal survival was investigated using Nissl staining at 14 days after MCAO. It was revealed that rTMS increased the neurological recovery of rats with MCAO, facilitated the migration of NSC, augmented the expression levels of the SDF-1α/CXCR4 axis and decreased neuronal loss. Furthermore, the rTMS-induced positive responses were significantly abolished by AMD3100. Overall, these results indicated that rTMS conferred therapeutic neuroprotective properties, which can restore neurological function after ischaemic stroke, in a manner that may be associated with the activation of the SDF-1α/CXCR4 axis.
Collapse
Affiliation(s)
- Yuguo Deng
- Department of Rehabilitation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Feng Guo
- Department of Rehabilitation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Xiaohua Han
- Department of Rehabilitation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Xiaolin Huang
- Department of Rehabilitation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| |
Collapse
|
12
|
Zhou L, Huang X, Li H, Guo R, Wang J, Zhang Y, Lu Z. Rehabilitation effect of rTMS combined with cognitive training on cognitive impairment after traumatic brain injury. Am J Transl Res 2021; 13:11711-11717. [PMID: 34786098 PMCID: PMC8581933] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Accepted: 08/15/2021] [Indexed: 12/08/2022]
Abstract
OBJECTIVE To innvestigate the rehabilitation effects of repetitive transcranial magnetic stimulation (rTMS) combined with cognitive training on cognitive impairment in patients with traumatic brain injury (TBI) by using multimodal magnetic resonance imaging. METHODS Clinical data of 166 patients with cognitive impairment after TBI were retrospectively analyzed. The patients were assigned into an observation group and a control group according to different treatment methods, with 83 cases in each group. The observation group was given rTMS + cognitive training, and the control group was given cognitive training only. The changes in GCS score, the Cho/Cr, Cho/NAA and NAA/Cr ratios examined by MRSI, the score of cognitive impairment, the grading of cognitive impairment, and the changes in modified Barthel index were observed and compared between the two groups. RESULTS The GCS score, and the ratios of Cho/Cr, Cho/NAA and NAA/Cr after treatment were better than those before treatment in both groups and were lower in the observation group compared with the control group (all P<0.05). The score and grading of cognitive impairment as well as modified Barthel index after treatment were all significantly better in the observation group than in the control group (all P<0.05). CONCLUSION rTMS can improve the rehabilitation effect on cognitive impairment in patients after TBI and is recommended for clinical use.
Collapse
Affiliation(s)
- Li Zhou
- Department of Rehabilitation, The First Affiliated Hospital of Guangdong Pharmaceutical UniversityGuangzhou, Guangdong Province, China
| | - Xuming Huang
- Department of Rehabilitation, The First Affiliated Hospital of Guangdong Pharmaceutical UniversityGuangzhou, Guangdong Province, China
| | - Haiyan Li
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-sen UniversityGuangzhou, Guangdong Province, China
| | - Ruomi Guo
- Department of Radiology, The Third Affiliated Hospital of Sun Yat-sen UniversityGuangzhou, Guangdong Province, China
| | - Jihui Wang
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-sen UniversityGuangzhou, Guangdong Province, China
| | - Yu Zhang
- Department of Pathology, Guangdong Provincial Hospital of Traditional Chinese MedicineGuangzhou, Guangdong Province, China
| | - Zhengqi Lu
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-sen UniversityGuangzhou, Guangdong Province, China
| |
Collapse
|
13
|
Wang F, Zhang C, Hou S, Geng X. Synergistic Effects of Mesenchymal Stem Cell Transplantation and Repetitive Transcranial Magnetic Stimulation on Promoting Autophagy and Synaptic Plasticity in Vascular Dementia. J Gerontol A Biol Sci Med Sci 2020; 74:1341-1350. [PMID: 30256913 DOI: 10.1093/gerona/gly221] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Indexed: 02/07/2023] Open
Abstract
Repetitive transcranial magnetic stimulation (rTMS) and mesenchymal stem cells (MSCs) transplantation both showed therapeutic effects on cognition impairment in vascular dementia (VD) model rats. However, whether these two therapies have synergistic effects and the molecular mechanisms remain unclear. In our present study, rats were randomly divided into six groups: control group, sham operation group, VD group, MSC group, rTMS group, and MSC+rTMS group. The VD model rats were prepared using a modified 2VO method. rTMS treatment was implemented at a frequency of 5 Hz, the stimulation intensity for 0.5 Tesla, 20 strings every day with 10 pulses per string and six treatment courses. The results of the Morris water maze test showed that the learning and memory abilities of the MSC group, rTMS group, and MSC+rTMS group were better than that of the VD group, and the MSC+rTMS group showed the most significant effect. The protein expression levels of brain-derived neurotrophic factor, NR1, LC3-II, and Beclin-1 were the highest and p62 protein was the lowest in the MSC+rTMS group. Our findings demonstrated that rTMS could further enhance the effect of MSC transplantation on VD rats and provided an important basis for the combined application of MSC transplantation and rTMS to treat VD or other neurological diseases.
Collapse
Affiliation(s)
- Fei Wang
- Department of Neurology, General Hospital, Tianjin Medical University, China
| | - Chi Zhang
- Department of Neurology, General Hospital, Tianjin Medical University, China
| | - Siyuan Hou
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Tianjin Medical University, China.,Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Tianjin Medical University, China
| | - Xin Geng
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Tianjin Medical University, China.,Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Tianjin Medical University, China
| |
Collapse
|
14
|
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: 21] [Impact Index Per Article: 4.2] [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
|
15
|
Leung A, Shirvalkar P, Chen R, Kuluva J, Vaninetti M, Bermudes R, Poree L, Wassermann EM, Kopell B, Levy R. Transcranial Magnetic Stimulation for Pain, Headache, and Comorbid Depression: INS-NANS Expert Consensus Panel Review and Recommendation. Neuromodulation 2020; 23:267-290. [PMID: 32212288 DOI: 10.1111/ner.13094] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Revised: 10/27/2019] [Accepted: 11/25/2019] [Indexed: 12/21/2022]
Abstract
BACKGROUND While transcranial magnetic stimulation (TMS) has been studied for the treatment of psychiatric disorders, emerging evidence supports its use for pain and headache by stimulating either motor cortex (M1) or dorsolateral prefrontal cortex (DLPFC). However, its clinical implementation is hindered due to a lack of consensus in the quality of clinical evidence and treatment recommendation/guideline(s). Thus, working collaboratively, this multinational multidisciplinary expert panel aims to: 1) assess and rate the existing outcome evidence of TMS in various pain/headache conditions; 2) provide TMS treatment recommendation/guidelines for the evaluated conditions and comorbid depression; and 3) assess the cost-effectiveness and technical issues relevant to the long-term clinical implementation of TMS for pain and headache. METHODS Seven task groups were formed under the guidance of a 5-member steering committee with four task groups assessing the utilization of TMS in the treatment of Neuropathic Pain (NP), Acute Pain, Primary Headache Disorders, and Posttraumatic Brain Injury related Headaches (PTBI-HA), and remaining three assessing the treatment for both pain and comorbid depression, and the cost-effectiveness and technological issues relevant to the treatment. RESULTS The panel rated the overall level of evidence and recommendability for clinical implementation of TMS as: 1) high and extremely/strongly for both NP and PTBI-HA respectively; 2) moderate for postoperative pain and migraine prevention, and recommendable for migraine prevention. While the use of TMS for treating both pain and depression in one setting is clinically and financially sound, more studies are required to fully assess the long-term benefit of the treatment for the two highly comorbid conditions, especially with neuronavigation. CONCLUSIONS After extensive literature review, the panel provided recommendations and treatment guidelines for TMS in managing neuropathic pain and headaches. In addition, the panel also recommended more outcome and cost-effectiveness studies to assess the feasibility of the long-term clinical implementation of the treatment.
Collapse
Affiliation(s)
- Albert Leung
- Professor of Anesthesiology and Pain Medicine, Department of Anesthesiology, Center for Pain Medicine, University of California, San Diego, School of Medicine, La Jolla, CA, USA.,Director, Center for Pain and Headache Research, VA San Diego Healthcare System, La Jolla, CA, USA
| | - Prasad Shirvalkar
- Assistant Professor, Departments of Anesthesiology (Pain Management), Neurology, and Neurosurgery, UCSF School of Medicine, USA
| | - Robert Chen
- Catherine Manson Chair in Movement Disorders, Professor of Medicine (Neurology), University of Toronto, Toronto, Ontario, Canada
| | - Joshua Kuluva
- Neurologist and Psychiatrist, TMS Health Solution, San Francisco, CA, USA
| | - Michael Vaninetti
- Assistant Clinical Professor, Anesthesiology and Pain Medicine, UCSD School of Medicine, La Jolla, CA, USA
| | - Richard Bermudes
- Chief Medical Officer, TMS Health Solutions, Assistant Clinical Professor- Volunteer, Department of Psychiatry, University of California, San Francisco, San Francisco, CA, USA
| | - Lawrence Poree
- Professor of Anesthesiology, Director, Neuromodulation Service, Division of Pain Medicine, University of California, San Francisco, School of Medicine, San Francisco, CA, USA
| | - Eric M Wassermann
- Director, Behavioral Neurology Unit, National Institute of Neurological Disorders and Stroke, Bethesda, MD, USA
| | - Brian Kopell
- Professor of Neurosurgery, Mount Sinai Center for Neuromodulation, New York, NY, USA
| | - Robert Levy
- President of International Neuromodulation Society, Editor-in-Chief, Neuromodulation, Boca Raton, FL, USA
| | -
- See Appendix for Complete List of Task Group Members
| |
Collapse
|
16
|
Zheng T, Yuan Y, Yang H, Du J, Wu S, Jin Y, Wang Z, Liu D, Shi Q, Wang X, Liu L. Evaluating the Therapeutic Effect of Low-Intensity Transcranial Ultrasound on Traumatic Brain Injury With Diffusion Kurtosis Imaging. J Magn Reson Imaging 2020; 52:520-531. [PMID: 31999388 DOI: 10.1002/jmri.27063] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Revised: 01/08/2020] [Accepted: 01/09/2020] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Low-intensity transcranial ultrasound (LITUS) has a therapeutic effect on traumatic brain injury (TBI). Diffusion kurtosis imaging (DKI) might be able to evaluate the effect changes of injured brain microstructure. PURPOSE To evaluate the therapeutic effect of LITUS in a moderate TBI rat model with DKI parameters. STUDY TYPE Prospective case-control animal study. ANIMAL MODEL Forty-five rats were randomly divided into sham control, TBI, and LITUS treatment groups (n = 15). FIELD STRENGTH/SEQUENCE Single-shot spin echo echo-planar imaging and fast T2 WI sequences at 3.0T. ASSESSMENT DKI parameters were obtained on days 1, 7, 14, 21, 28, 35, and 42 after TBI. STATISTICAL TESTS For the mean kurtosis (MK), axial kurtosis (Ka), and radial kurtosis (Kr) values, groups were compared using a two-way analysis of variance (ANOVA). RESULTS LITUS inhibited TBI and caused MK values to increase significantly during the early stage (LITUS vs. TBI, day 7, adjusted P < 0.0001) and decrease during the late stage (LITUS vs. TBI, day 42, adjusted P = 0.0156) in the damaged cortex. In the thalamus, the MK value of the TBI group began to rise on day 7, with no change observed in the LITUS group. TBI increases Ka value during the early stage in the cortex and decreases during the late stage in the cortex and thalamus. LITUS inhibited these Ka changes (LITUS vs. TBI, day 7, adjusted P = 0.0014; LITUS vs. TBI, day 42, adjusted P = 0.0026 and 0.0478, respectively, for cortex and thalamus). The Kr value increased slightly during the early stage in the cortex (TBI vs. Sham, day 1, adjusted P = 0.0016). DATA CONCLUSION The DKI parameter, particularly the MK value, evaluates primary cortical injury as well as the secondary brain injury that could not be detected by conventional T2 WI. LEVEL OF EVIDENCE 1 Technical Efficacy Stage: 4 J. Magn. Reson. Imaging 2020;52:520-531.
Collapse
Affiliation(s)
- Tao Zheng
- Department of Magnetic Resonance Imaging, Qinhuangdao Municipal No. 1 Hospital, Qinhuangdao, China
| | - Yi Yuan
- Institute of Electrical Engineering, Yanshan University, Qinhuangdao, China
| | - Haoxiang Yang
- Department of Cardiovascular Medicine, Qinhuangdao Municipal No. 1 Hospital, Qinhuangdao, China
| | - Juan Du
- Department of Magnetic Resonance Imaging, Qinhuangdao Municipal No. 1 Hospital, Qinhuangdao, China
| | - Shuo Wu
- Department of Magnetic Resonance Imaging, Qinhuangdao Municipal No. 1 Hospital, Qinhuangdao, China
| | - Yinglan Jin
- Peking University Health Science Center, Beijing, China
| | - Zhanqiu Wang
- Department of Magnetic Resonance Imaging, Qinhuangdao Municipal No. 1 Hospital, Qinhuangdao, China
| | - Defeng Liu
- Department of Magnetic Resonance Imaging, Qinhuangdao Municipal No. 1 Hospital, Qinhuangdao, China
| | - Qinglei Shi
- Scientific Clinical Specialist, Siemens Ltd., Beijing, China
| | - Xiaohan Wang
- Department of Magnetic Resonance Imaging, Qinhuangdao Municipal No. 1 Hospital, Qinhuangdao, China
| | - Lanxiang Liu
- Department of Magnetic Resonance Imaging, Qinhuangdao Municipal No. 1 Hospital, Qinhuangdao, China
| |
Collapse
|
17
|
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
|
18
|
Rashno M, Sarkaki A, Farbood Y, Rashno M, Khorsandi L, Naseri MKG, Dianat M. Therapeutic effects of chrysin in a rat model of traumatic brain injury: A behavioral, biochemical, and histological study. Life Sci 2019; 228:285-294. [PMID: 31063733 DOI: 10.1016/j.lfs.2019.05.007] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Revised: 04/28/2019] [Accepted: 05/03/2019] [Indexed: 12/20/2022]
Abstract
AIMS Oxidative stress and apoptosis have major roles in the progression of traumatic brain injury (TBI)-associated motor and cognitive deficits. The present study was aimed to elucidate the putative effects of chrysin, a natural flavonoid compound, against TBI-induced motor and cognitive dysfunctions and possible involved mechanisms. MAIN METHODS Chrysin (25, 50 or 100 mg/kg) was orally administered to rats starting immediately following TBI induction by Marmarou's weight-drop technique and continuously for 3 or 14 days. Neurological functions, motor coordination, learning and memory performances, histological changes, cell apoptosis, expression of pro- and anti-apoptotic proteins, and oxidative status were assayed at scheduled time points after experimental TBI. KEY FINDINGS The results indicated that treatment with chrysin improved learning and memory disabilities in passive avoidance task, and ameliorated motor coordination impairment in rotarod test after TBI. These beneficial effects were accompanied by increased the concentrations of superoxide dismutase (SOD), catalase (CAT), glutathione peroxidase (GPx), glutathione (GSH), decreased malondialdehyde (MDA) content, prevented neuronal loss, diminished apoptotic index, elevated the expression of anti-apoptotic Bcl-2 protein, and reduced the expression of pro-apoptotic Bax protein in the cerebral cortex and hippocampus tissues. SIGNIFICANCE Our findings suggest that both anti-oxidative and anti-apoptotic properties of chrysin (especially in the dose of 100 mg/kg) are possible mechanisms that improve cognitive/motor deficits and prevent neuronal cell death after TBI.
Collapse
Affiliation(s)
- Masome Rashno
- Department of Physiology, Faculty of Medicine, Physiology Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Alireza Sarkaki
- Department of Physiology, Faculty of Medicine, Physiology Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
| | - Yaghoob Farbood
- Department of Physiology, Faculty of Medicine, Physiology Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Mohammad Rashno
- Department of Immunology, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran; Cellular and Molecular Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Layasadat Khorsandi
- Cellular and Molecular Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Mohammad Kazem Gharib Naseri
- Department of Physiology, Faculty of Medicine, Physiology Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Mahin Dianat
- Department of Physiology, Faculty of Medicine, Physiology Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| |
Collapse
|
19
|
Martens KM, Pechacek KM, Modrak CG, Milleson VJ, Zhu B, Vonder Haar C. Cathodal Transcranial Direct-Current Stimulation Selectively Decreases Impulsivity after Traumatic Brain Injury in Rats. J Neurotrauma 2019; 36:2827-2830. [PMID: 31072218 PMCID: PMC6744944 DOI: 10.1089/neu.2019.6470] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Traumatic brain injury (TBI) often results in chronic psychiatric-like symptoms. In a condition with few therapeutic options, neuromodulation has emerged as a promising potential treatment avenue for these individuals. The goal of the current study was to determine if transcranial direct-current stimulation (tDCS) could treat deficits of impulsivity and attention in rats. This could then be used as a model to investigate treatment parameters and the mechanism of action underlying therapeutic effects. Rats were trained on a task to measure attention and motor impulsivity (five-choice serial reaction time task), then given a frontal, controlled cortical impact injury. After rats recovered to a new baseline, tDCS (cathodal, 10 min, 800 μA) was delivered daily prior to testing in a counterbalanced, cross-over design. Treatment with tDCS selectively reduced impulsivity in the TBI group, and the greatest recovery occurred in the rats with the largest deficits. With these data, we have established a rat model for studying the effects of tDCS on psychiatric-like dysfunction. More research is needed to determine the mechanism of action by which tDCS-related gains occur.
Collapse
Affiliation(s)
- Kris M Martens
- Injury and Recovery Laboratory, Department of Psychology, West Virginia University, Morgantown, West Virginia.,Department of Neuroscience, West Virginia University, Morgantown, West Virginia
| | - Kristen M Pechacek
- Injury and Recovery Laboratory, Department of Psychology, West Virginia University, Morgantown, West Virginia
| | - Cassandra G Modrak
- Injury and Recovery Laboratory, Department of Psychology, West Virginia University, Morgantown, West Virginia
| | - Virginia J Milleson
- Injury and Recovery Laboratory, Department of Psychology, West Virginia University, Morgantown, West Virginia
| | - Binxing Zhu
- Injury and Recovery Laboratory, Department of Psychology, West Virginia University, Morgantown, West Virginia
| | - Cole Vonder Haar
- Injury and Recovery Laboratory, Department of Psychology, West Virginia University, Morgantown, West Virginia.,Department of Neuroscience, West Virginia University, Morgantown, West Virginia
| |
Collapse
|
20
|
Long J, He C, Dai H, Kang X, Zou J, Ye S, Yu Q. Effects of transection of cervical sympathetic trunk on cognitive function of traumatic brain injury rats. Neuropsychiatr Dis Treat 2019; 15:1121-1131. [PMID: 31118645 PMCID: PMC6506013 DOI: 10.2147/ndt.s199450] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/25/2018] [Accepted: 03/28/2019] [Indexed: 01/04/2023] Open
Abstract
Objective: To observe the effects of transection of cervical sympathetic trunk (TCST) on the cognitive function of traumatic brain injury (TBI) rats and the potential mechanisms. Methods: A total of 288 adult male SD rats were divided into 3 groups using a random number table: TBI group (n=96), TBI + TCST group (n=96) and Sham group (n=96). The water maze test was performed before TBI (T0) and at day 1 (T1), day 2 (T2), day 3 (T3), 1 week (T4), 2 weeks (T5), 6 weeks (T6) and 12 weeks (T7) after TBI. The levels of α1-adrenergic receptors (α1-ARs), α2-adrenergic receptors (α2-ARs), toll-like receptor 4 (TLR-4) and P38 in hippocampi were detected by real-time PCR. Hippocampal P38 expression was assayed by Western blot. The expressions of interleukin-6 (IL-6), tumor necrosis factor (TNF-α) and brain-derived neurotrophic factor (BDNF) were examined by immunohistochemistry. Noradrenaline (NE) expression in plasma was evaluated by ELISA. The respiratory control ratio (RCR) of brain mitochondria was detected using a Clark oxygen electrode. Results: TCST effectively improved the cognitive function of TBI rats. TCST significantly inhibited sympathetic activity in the rats and effectively inhibited inflammatory responses. The expression of BDNF at T1-T6 in TBI+TCST group was higher than that in TBI group (P<0.05). Furthermore, P38 expression was inhibited more effectively in TBI+TCST group (P<0.05), than in TBI group (P<0.05), and the RCR of the brain was significantly higher in TBI+TCST group than in TBI group (P<0.05). Conclusions: TCST can enhance cognitive function in TBI rats by inhibiting sympathetic activity, reducing inflammatory responses and brain edema, upregulating BDNF and improving brain mitochondrial function.
Collapse
Affiliation(s)
- Juan Long
- Department of Pain, Guizhou Province People's Hospital, Guiyang, People's Republic of China
| | - Chunjing He
- Department of Pain, Guizhou Province People's Hospital, Guiyang, People's Republic of China
| | - Hong Dai
- Department of Neurology, Guizhou Province People's Hospital, Guiyang, People's Republic of China
| | - Xinguo Kang
- Department of Pain, Guizhou Province People's Hospital, Guiyang, People's Republic of China
| | - Jinfeng Zou
- Department of Pain, Guizhou Province People's Hospital, Guiyang, People's Republic of China
| | - Shengli Ye
- Department of Pain, Guizhou Province People's Hospital, Guiyang, People's Republic of China
| | - Qian Yu
- Department of Neurology, Guizhou Province People's Hospital, Guiyang, People's Republic of China
| |
Collapse
|
21
|
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
|
22
|
Duan X, Yao G, Liu Z, Cui R, Yang W. Mechanisms of Transcranial Magnetic Stimulation Treating on Post-stroke Depression. Front Hum Neurosci 2018; 12:215. [PMID: 29899693 PMCID: PMC5988869 DOI: 10.3389/fnhum.2018.00215] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Accepted: 05/08/2018] [Indexed: 12/13/2022] Open
Abstract
Post-stroke depression (PSD) is a neuropsychiatric affective disorder that can develop after stroke. Patients with PSD show poorer functional and recovery outcomes than patients with stroke who do not suffer from depression. The risk of suicide is also higher in patients with PSD. PSD appears to be associated with complex pathophysiological mechanisms involving both psychological and psychiatric problems that are associated with functional deficits and neurochemical changes secondary to brain damage. Transcranial magnetic stimulation (TMS) is a non-invasive way to investigate cortical excitability via magnetic stimulation of the brain. TMS is currently a valuable tool that can help us understand the pathophysiology of PSD. Although repetitive TMS (rTMS) is an effective treatment for patients with PSD, its mechanism of action remains unknown. Here, we review the known mechanisms underlying rTMS as a tool for better understanding PSD pathophysiology. It should be helpful when considering using rTMS as a therapeutic strategy for PSD.
Collapse
Affiliation(s)
- Xiaoqin Duan
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetics, The Second Hospital of Jilin University, Changchun, China
| | - Gang Yao
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetics, The Second Hospital of Jilin University, Changchun, China
| | - Zhongliang Liu
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetics, The Second Hospital of Jilin University, Changchun, China
| | - Ranji Cui
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetics, The Second Hospital of Jilin University, Changchun, China
| | - Wei Yang
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetics, The Second Hospital of Jilin University, Changchun, China
| |
Collapse
|
23
|
Zhang K, Shi Z, Zhou J, Xing Q, Ma S, Li Q, Zhang Y, Yao M, Wang X, Li Q, Li J, Guan F. Potential application of an injectable hydrogel scaffold loaded with mesenchymal stem cells for treating traumatic brain injury. J Mater Chem B 2018; 6:2982-2992. [DOI: 10.1039/c7tb03213g] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
In this contribution, we developed an injectable hydrogel composed of sodium alginate and hyaluronic acid that acts as a tissue scaffold to create a more optimal microenvironment for the stem cells for potential application of traumatic brain injury implantation.
Collapse
|